WO2011028952A1 - Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens - Google Patents

Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens Download PDF

Info

Publication number
WO2011028952A1
WO2011028952A1 PCT/US2010/047741 US2010047741W WO2011028952A1 WO 2011028952 A1 WO2011028952 A1 WO 2011028952A1 US 2010047741 W US2010047741 W US 2010047741W WO 2011028952 A1 WO2011028952 A1 WO 2011028952A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
domain
antigen
mab
binding
Prior art date
Application number
PCT/US2010/047741
Other languages
French (fr)
Inventor
Gregory A. Lazar
Gregory L. Moore
Original Assignee
Xencor, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Xencor, Inc. filed Critical Xencor, Inc.
Publication of WO2011028952A1 publication Critical patent/WO2011028952A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against Fc-receptors, e.g. CD16, CD32, CD64
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • novel antibody analogs described preferably utilize heterodimeric Fc regions. Methods of using the novel antibody analog compositions, particularly for therapeutic purposes, are also described herein.
  • Antibody-based therapeutics have been used successivefully to treat a variety of diseases, including cancer and autoimmune/inflammatory disorders. Yet improvements to this class of drugs are still needed, particularly with respect to enhancing their clinical efficacy.
  • One avenue being explored is the engineering of additional and novel antigen binding sites into antibody-based drugs such that a single immunoglobulin molecule co- engages two different antigens. Such non-native or alternate antibody formats that engage two different antigens are often referred to as bispecifics. Because the considerable diversity of the antibody variable region (Fv) makes it possible to produce an Fv that recognizes virtually any molecule, the typical approach to bispecific generation is the introduction of new variable regions into the antibody.
  • Fv antibody variable region
  • bispecific antibodies were made by fusing two cell lines that each produced a single monoclonal antibody (Milstein et al., 1983, Nature 305:537-540). Although the resulting hybrid hybridoma or quadroma did produce bispecific antibodies, they were only a minor population, and extensive purification was required to isolate the desired antibody. An engineering solution to this was the use of antibody fragments to make bispecifics.
  • variable light and heavy chains can be linked in single genetic constructs.
  • Antibody fragments of many different forms have been generated, including diabodies, single chain diabodies, tandem scFv's, and Fab 2 bispecifics (Chames & Baty, 2009, mAbs 1 [6]:1 -9; Holliger & Hudson, 2005, Nature
  • a principal cause of these drawbacks is that antibody fragments typically lack the constant region of the antibody with its associated functional properties, including larger size, high stability, and binding to various Fc receptors and ligands that maintain long half-life in serum (i.e. the neonatal Fc receptor FcRn) or serve as binding sites for purification (i.e. protein A and protein G).
  • the desired binding is monovalent rather than bivalent.
  • cellular activation is accomplished by cross-linking of a monovalent binding interaction.
  • the mechanism of cross-linking is typically mediated by antibody/antigen immune complexes, or via effector cell to target cell engagement.
  • FcyRs the low affinity Fc gamma receptors
  • FcyRs such as FcyRlla, FcyRllb, and FcyRllla bind monovalently to the antibody Fc region.
  • T-cells a mechanism that is mediated by immune complexation of soluble IgG's with the same antigen that is recognized by the BCR (Heyman 2003, Immunol Lett 88[2]:157-161 ; Smith and Clatworthy, 2010, Nature Reviews Immunology 10:328-343; expressly incorporated by reference).
  • CD3 activation of T-cells occurs only when its associated T- cell receptor (TCR) engages antigen-loaded MHC on antigen presenting cells in a highly avid cell-to-cell synapse (Kuhns et al., 2006, Immunity 24:133-139).
  • the present disclosure provides novel antibody analog compositions that co-engage a first and second antigen.
  • FIG. 1 Heterodimeric Fc region proof-of-concept system.
  • DNA constructs encoding two different immunoglobulin polypeptides were designed: (i) scFv-Fc (VH-linker-VK-Hinge- CH2-CH3') and (ii) empty-Fc (Hinge-CH2-CH3").
  • scFv-Fc VH-linker-VK-Hinge- CH2-CH3'
  • ii empty-Fc
  • three possible dimers can form from cotransfection and expression of these two polypeptides, each composed of a different number of immunoglobulin (Ig) domains and thus different molecular weights.
  • the two different CH3 domains, CH3' (prime) and CH3" (double prime) can be designed to promote heterodimer formation and discourage homodimer formation.
  • FIG. 1 Amino acid sequences of heterodimer constructs and native lgG1 and lgG2 constant heavy chains (SEQ ID NOs. 1-4).
  • Figure 3. Electrophoretic visualization of protein species generated by simultaneous expression of empty-Fc and scFv-Fc polypeptides (each with wild-type lgG1 Fc sequence) using Agilent 2100 Bioanalyzer microfluidics-based platform. The molecular weights of the proteins in the ladder lane (designated L) are shown on the left in kilodaltons (KDa).
  • Lanes 1-5 reducing environment, cotransfection DNA ratios of empty-Fc:scFv-Fc 1 :1 , 1 :2, 1 :4, 2:1 , 4:1 ; Lanes 6-10: non-reducing
  • the top band (-70 KDa) is scFv-Fc monomer
  • the bottom band (-38 kDa) is empty-Fc monomer.
  • the top band (-134 KDa) is scFv-Fc homodimer
  • the middle band (-105 KDa) is scFc-Fv/empty-Fc heterodimer
  • the bottom band (-72 KDa) is empty-Fc homodimer.
  • the molecular weights of the proteins in the ladder lane (designated L) are shown on the left in kilodaltons (KDa).
  • KDa kilodaltons
  • FIG. Protein species by molar % from simultaneous expression of variant empty- Fc and variant scFv-Fc.
  • Empty-Fc refers to substitutions in the Empty-Fc chain and scFv-Fc refers to substitutions in the scFv-Fc chain.
  • FIG. 7 Protein species by mass % from simultaneous expression of variant empty- Fc and variant scFv-Fc.
  • Empty-Fc refers to substitutions in the Empty-Fc chain and scFv-Fc refers to substitutions in the scFv-Fc chain.
  • FIG. 8 Illustration of mAb-Fv and mAb-Fab immunoglobulin formats.
  • a native lgG1 antibody is included for reference.
  • mAb-Fv and mAb-Fab immunoglobulins can be constructed with homodimeric CH3 domains, preferably they utilize heterodimeric CH3 domains, as depicted.
  • Bivalent binding to antigen-1 (Ag-1 ) is mediated by the N-terminal VH/VL pairs (Fv-1 ), while monovalent binding to antigen-2 (Ag-2) is mediated by the C- terminal VH/VL pair (Fv-2).
  • Fab-Fv and Fab-Fab analogs that bind both antigen-1 and antigen-2 monovalently.
  • FIG. 9 Sequences of mAb-Fv constructs with Fv-2 targeting FcYRIIIa.
  • LC refers to light chain
  • HC1 and HC2 refer to the two heavy chains that make up each mAb-Fv.
  • Identification (ID) numbers correspond to those in Table 3 and the results.
  • 9064 is a mAb- Fab, all other constructs are mAb-Fv's.
  • the linker separating the Fc region with Fv-2 is underlined, and Fv-2 and any C-terminal linkers are shown in italics.
  • the histidine tags in enumerated sequences are optional, and generally not included in therapeutic compositions (SEQ ID NOs. 5-21 ).
  • FIG. 10 Electrophoretic visualization of mAb-Fvs and mAb-Fab with Fv-2 targeting FcYRIIIa using the Agilent Bioanalyzer. Environment is non-reducing (“nr") or reducing ("red”) as labeled. The molecular weights of the proteins in the ladder lane (L) are shown on the left in kilodaltons (KDa).
  • Figure 1 1 Size exclusion chromatograms for aFcYRIIIa co-engaging mAb-Fv's 9516, 9517, and 9518.
  • FIG. 12 Biacore® sensorgrams depicting the binding of various FCYRS to mAb-Fv and antibodies captured by a protein A chip. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer).
  • FIG. 13 Biacore® sensorgrams depicting the binding of FcRn to mAb-Fv and antibodies captured by a HER2 chip. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer).
  • FIG. 14 Biacore® sensorgrams depicting the binding of various concentrations of FcYRIIIa(V158 isoform) to protein A-captured 9516 mAb-Fv. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer). The K d of 1 1 1 nM was calculated by global fitting of binding data with a 1 :1 Langmuir binding model using BIAevaluation software.
  • FIG. 15 Biacore® sensorgrams depicting the simultaneous co-engagement of Her2 and Fc Rllla by mAb-Fv. 9516 (aHER2 x aFcYRIIIa) was captured to a HER2 chip followed by binding of FcYRII Ia(158V). Data were processed by zeroing time and response before the injection of 9516.
  • FIG. 1 Biacore sensorgrams depicting the capture of 9518 ( ⁇ 1 .24 x aFcYRII Ia) by a HM 1 .24 chip followed by dual binding of FcYRI I Ia(158V). Data were processed by zeroing time and response before the injection of 9518.
  • FIG. 1 Cytotoxicity of various mAb-Fvs and mAbs against SK-BR-3 cells using human PBMCs. FcYRI IIa-dependent cytotoxicity was measured at multiple mAb-Fv or mAb concentrations by lactate dehydrogenase release (mean ⁇ SE of triplicate wells).
  • FIG. 20 Electrophoretic visualization of mAb-Fvs with Fv-2 targeting FcYRI Ib using the Agilent Bioanalyzer. Environment is non-reducing (“nr") or reducing ("red”) as labeled. The molecular weights of the proteins in the ladder lane (L) are shown on the left in kilodaltons (KDa).
  • FIG. 21 Biacore® sensorgrams depicting the binding of various concentrations of FcYRI Ib to protein A-captured 9547 mAb-Fv. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer). The K d of 44 nM was calculated by global fitting of binding data with a 1 : 1 Langmuir binding model using
  • FIG. 22 Calcium response of CD40+ PBMCs after incubation with various concentrations of 9547 (aCD19 x aFcYRI Ib) or 6187 (aCD19 FcKO). Calcium mobilization was induced with 10 [ig/m ⁇ anti-CD79b (G236R/L328R) BCR-activating antibody.
  • FIG. 24 Electrophoretic visualization of mAb-Fvs with Fv-2 targeting CD3 using the Agilent Bioanalyzer. Environment is non-reducing (“nr") or reducing ("red”) as labeled. The molecular weights of the proteins in the ladder lane (L) are shown on the left in kilodaltons (KDa).
  • FIG. 25 Size exclusion chromatograms for aCD3 co-engaging mAb-Fvs.
  • Figure 26 Competion of unlabeled 9509 aHER2 x aCD3 mAb-Fv with FITC-labeled full-length OKT3 mAb for binding to CD3+ T cells in PBMCs.
  • FIG. 27 Cytotoxicity of mAb-Fvs against Her2+ SK-BR-3 cells using T cells isolated from human PBMCs. CD3-dependent cytotoxicity was measured at multiple mAb-Fv concentrations by lactate dehydrogenase release (mean ⁇ SE of triplicate wells).
  • FIG. 28 Cytotoxicity of mAb-Fvs against Her2+ SKOV3 cells using T cells isolated from human PBMCs. CD3-dependent cytotoxicity was measured at multiple mAb-Fv concentrations by lactate dehydrogenase release (mean ⁇ SE of triplicate wells).
  • FIG. 29 Cytotoxicity of various mAb-Fvs and mAbs against HM1 .24+ RPMI 8224 cells using human PBMCs. CD3-dependent cytotoxicity was measured at multiple mAb-Fv or mAb concentrations by lactate dehydrogenase release (mean ⁇ SE of triplicate wells).
  • the present invention is directed to a variety of antibodies (generally referred to herein as "antibody analogs", as further described below) that co-engage at least two different antigens in novel ways.
  • the two antigens of the invention are generally referred to as antigen-1 and antigen-2 (with some embodiments, described below, including antigen-3 as a third antigen).
  • antigen-1 and antigen-2 with some embodiments, described below, including antigen-3 as a third antigen.
  • antigen-3 as a third antigen.
  • there are two different types of antibody analogs that allow for co-engagement mechanisms one that utilizes three antigen binding domains (e.g. one antigen is bound bivalently and the other is bound monovalently, although as is further described below, there can also be three different antigens that are bound or a single antigen), and one that relies on two antigen binding domains (e.g. each antigen is bound monovalently).
  • antibody analogs are made that have three antigen binding domains.
  • This embodiment can be exemplified in a variety of ways. In an embodiment of particular use, this is done by using a "traditional" antibody structure (two heavy chains with associated light chains that form antigen binding domains) that includes an additional antigen binding domain at the "opposite end" of the antibody. That is, in some
  • all three of the antigen binding domains of the antibody analog are antibody variable regions, comprising a variable heavy and variable light chain.
  • two of the antigen binding domains bind to antigen-1 , e.g. bivalently, and the third antigen binding domain binds monovalently to antigen-2.
  • monovalent binding can be preferred, to avoid cross-linking and/or activation of certain pathways.
  • this "trimeric binding domain" embodiment can be
  • heterodimeric heavy chains differ from one another in one or more ways.
  • one of the heavy chains has a C-terminal fusion (e.g. fused to the C-terminus of the CH3 domain of the first heavy chain) that comprises an "extra” (sometimes referred to herein as “exogenous") variable heavy domain.
  • the other heavy chain also has a C-terminal fusion that comprises an exogenous variable light domain.
  • the two "extra" variable domains form an antigen binding domain.
  • these antibody analogs have a third binding domain.
  • the two heavy chains also differ from each other by the presence of amino acid substitutions in the Fc domain that favors heterodimeric formation over homodimers. That is, by changing amino acids in each heavy chain, different heavy chains are more likely to associate to form the antibody analog structure than to form homodimers with the same Fc amino acid sequences.
  • antibody analogs are made that have two different antigen binding domains (although as described below, the antigens could be identical). While these antibody analogs share some functional similarity to bispecific antibodies and some antibody fragment constructs, the bispecific antibody analogs of the present invention are easier to make and use than traditional bispecific antibodies.
  • the "dimeric binding domain" antibody analog also has components of traditional antibodies.
  • the first heavy chain is identical to a "traditional" antibody construct, in that in contains the variable heavy and light regions, with the exception that it contains a C-terminal fusion at the CH3 domain of an exogenous variable domain (either heavy or light).
  • the second heavy chain comprises the Fc region of the heavy chain and an N-terminal variable light chain instead of a N-terminal variable heavy chain, such that the N-terminal portion of the analog forms a first antigen binding site (e.g. binds to antigen- 1 ).
  • the second heavy chain also contains a C-terminal fusion to the CH3 domain of an exogeneous variable domain, either heavy or light, such that one chain contains the exogenous heavy chain and the other the exogenous light chain.
  • This general scheme is shown in Figure 8.
  • the antigen binding regions of the antibody analog are antibody variable regions.
  • binding to antigens is mediated by variable regions, also referred to as Fv regions, each comprising a VH domain and a VL domain.
  • Fv-1 variable regions
  • Fv-2 variable regions
  • the antibody analog is a novel antibody format referred to as mAb-Fv or mAb-Fab.
  • mAb-Fv comprises an IgG antibody with a C-terminal Fv fusion.
  • mAb-Fab comprises an IgG antibody with a C- terminal Fab fusion.
  • the N-terminal Fab regions of the mAb-Fv and mAb-Fab contain Fv-1 . Because there are two such Fv-1 regions, the mAb-Fv and mAb-Fab bind bivalently to antigen-1 . That is, one immunoglublin binds two of antigen-1 at the same time.
  • the C-terminal Fv region of the mAb-Fv or C-terminal Fab region of the mAb-Fab contain Fv- 2, which is the binding site for antigen-2. Because there is only one such Fv-2 region, the mAb-Fv and mAb-Fab bind monovalently to antigen-2. That is, one immunoglobulin binds only one of antigen-2.
  • the mAb-Fv and mAb-Fab immunoglobulins thus contain three variable regions, two of which are Fv-1 and one of which is Fv-2.
  • the mAb-Fab differs from the mAb-Fv in that the former contains CH and CL constant domains C-terminal to the Fv-2 VH and VL domains respectively.
  • the mAb-Fv and mAb-Fab immunoglobulins comprise three distinct immunoglobulin chains. These chains include an antibody light chain and two antibody heavy chains. One of said heavy chains comprises a VH or VH-CH 1 at its C- terminus, whereas the other of said heavy chains comprises a VL or VL-CL at its C-terminus.
  • Fab-Fv and Fab-Fab are analogues of the mAb-Fv and mAb-Fab that bind both antigen-1 and antigen-2 monovalently. These immunoglobulins are referred to as Fab-Fv and Fab-Fab.
  • the Fab-Fv and Fab-Fab immunoglobulins comprise an N-terminal Fab region that contains a single binding site for antigen-1 , and comprise a C-terminal Fv or Fab region that contains a single binding site for antigen-2.
  • mAb-Fv and mAb-Fab immunoglobulins of the invention may also comprise single domain VHH regions as Fv-1 . That is, the N-terminal binding portion of the mAb-Fv and mAb-Fab may lack a light chain and a CH 1 domain.
  • the first and second antigens are distinct. That is, Fv-1 and Fv-2 bind different antigens. However, in some embodiments, antigen-1 and antigen-2 may be the same, i.e. Fv-1 and Fv-2 have specificity for the same antigen.
  • Fv-1 is replaced with a fusion protein, for example a receptor, that binds antigen-1.
  • a fusion protein for example a receptor
  • antigen-1 is bound by a fusion protein
  • antigen-2 is bound by Fv-2.
  • the novel antibody analogs described preferably comprise a heterodimeric Fc region.
  • a heterodimer Fc region, as described herein, is a pair of variant Fc regions that comprise at least one amino acid modification in each Fc region.
  • said amino acid modifications stabilize a heterodimeric Fc region while destabilizing homodimeric Fc regions.
  • said variant Fc regions comprise at least one substitution at a position selected from the group consisting of 349, 351 , 354, 356, 357, 364, 366, 368, 370, 392, 394, 395, 396, 397, 399, 401 , 405, 407, 409, 41 1 , and 439, wherein numbering is according to the EU index as in Kabat.
  • said variant Fc regions comprise at least one substitution selected from the group consisting of 349A, 349C, 349E, 349I, 349K, 349S, 349T, 349W, 351 E, 351 K, 354C, 356K, 357K, 364C, 364D, 364E, 364F, 364G, 364H, 364R, 364T, 364Y, 366D, 366K, 366S, 366W, 366Y, 368A, 368E, 368K, 368S, 370C, 370D, 370E, 370G, 370R, 370S, 370V, 392D, 392E, 394F, 394S, 394W, 394Y, 395T, 395V, 396T, 397E, 397S, 397T, 399K, 401 K, 405A, 405S, 407T, 407V, 409D, 409E,
  • the present invention describes methods for generating the novel compositions of the invention.
  • the present invention describes purification methods for the immunogloublins herein, particularly methods for separating heterodimeric and homodimeric protein species. Also described are methods of testing the immunogloublins herein, including in vitro and in vivo experiments.
  • the present invention provides isolated nucleic acids encoding the novel antibody analog compositions described herein.
  • the present invention provides vectors comprising said nucleic acids, optionally, operably linked to control sequences.
  • the present invention provides host cells containing the vectors, and methods for producing and optionally recovering the antibody analog compositions.
  • compositions comprising antibody analog
  • polypeptides described herein and a physiologically or pharmaceutically acceptable carrier or diluent.
  • present invention contemplates therapeutic and diagnostic uses for the antibody analog polypeptides disclosed herein.
  • amino acid and “amino acid identity” as used herein is meant one of the 20 naturally occurring amino acids or any non-natural analogues that may be present at a specific, defined position.
  • amino acid as used herein means both naturally occurring and synthetic amino acids. For example, homophenylalanine, citrulline and noreleucine are considered amino acids for the purposes of the invention.
  • Amino acid also includes imino acid residues such as proline and hydroxyproline.
  • the side chain may be in either the (R) or the (S) configuration. In the preferred embodiment, the amino acids are in the (S) or L- configuration. If non-naturally occurring side chains are used, non-amino acid substituents may be used, for example to prevent or retard in vivo degradation.
  • constant region of an antibody as defined herein is meant the region of the antibody that is encoded by one of the light or heavy chain immunoglobulin constant region genes.
  • constant light chain or constant region or “CL” as used herein is meant the region of an antibody encoded by the kappa (CK) or lambda (C ) light chains.
  • the constant light chain typically comprises a single domain, and as defined herein refers to positions 1 08-214 of CK or C , wherein numbering is according to the EU index.
  • constant heavy chain or “heavy chain constant region” as used herein is meant the region of an antibody encoded by the mu, delta, gamma, alpha, or epsilon genes to define the antibody's isotype as IgM, IgD, IgG, IgA, or IgE, respectively.
  • the constant heavy chain refers to the N-terminus of the CH 1 domain to the C-terminus of the CH3 domain, thus comprising positions 1 1 8-447, wherein numbering is according to the EU index.
  • Fab or "Fab region” as used herein is meant the polypeptides that comprise the VH, CH 1 , VL, and CL immunoglobulin domains. Fab may refer to this region in isolation, or this region in the context of a full length antibody or antibody fragment.
  • Fc or “Fc region”, as used herein is meant the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain.
  • Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains.
  • IgA and IgM Fc may include the J chain.
  • Fc comprises immunoglobulin domains Cgamma2 and Cgamma3 (Cy2 and Cy3) and the hinge between Cgammal (Cy1 ) and Cgamma2 (Cy2).
  • Fc region may vary, the human IgG heavy chain Fc region is usually defined to comprise residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat.
  • Fc may refer to this region in isolation, or this region in the context of an Fc polypeptide, for example an antibody.
  • Fc polypeptide as used herein is meant a polypeptide that comprises all or part of an Fc region.
  • Fc polypeptides include antibodies, Fc fusions, isolated Fes, and Fc fragments.
  • Fc fusion or “immunoadhesin” as used herein is meant a protein wherein one or more polypeptides is operably linked to an Fc domain.
  • Fc fusion is herein meant to be synonymous with the terms “immunoadhesin”, “Ig fusion”, “Ig chimera”, and “receptor globulin” as used in the prior art (Huang, 2009, Curr Opin Biotechnology 20:692-699;
  • An Fc fusion combines the Fc region of an immunoglobulin with a fusion partner, which in general may be any protein, polypeptide or small molecule.
  • a fusion partner which in general may be any protein, polypeptide or small molecule.
  • the role of the non-Fc part of an Fc fusion, i.e., the fusion partner, is to mediate target binding, and thus it is functionally analogous to the variable regions of an antibody.
  • Virtually any protein or small molecule may be linked to Fc to generate an Fc fusion.
  • Protein fusion partners may include, but are not limited to, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain.
  • Small molecule fusion partners may include any therapeutic agent that directs the Fc fusion to a therapeutic target. The role of the latter is to mediate target recognition, and thus it is functionally analogous to the antibody variable region. Because of the structural and functional overlap of Fc fusions with antibodies, the discussion on antibodies in the present disclosure extends also to Fc fusions.
  • Protein fusion partners may include, but are not limited to, the variable region of any antibody, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain.
  • Small molecule fusion partners may include any agent that directs the Fc fusion to a target antigen.
  • target antigen may be any molecule, e.g., an extracellular receptor, that is implicated in disease.
  • Fusion partners may be linked to any region of an Fc region, including at the N- or C- termini, or at some residue in-between the termini.
  • a fusion partner is linked at the N- or C-terminus of the Fc region.
  • Fv region or “variable region” when used herein refers to the minimum fragment of an antibody that retains both antigen-recognition and antigen-binding sites.
  • heterodimeric Fc region as used herein is meant an Fc region composed of two different heavy constant chains, e,g, having different sequences .
  • the changes can comprise one or more amino acid changes.
  • immunoglobulin (Iq) as used herein is meant a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. Immunoglobulins include but are not limited to antibodies. Immunoglobulins may have a number of structural forms, including but not limited to full length antibodies, antibody fragments, and individual immunoglobulin domains.
  • immunoglobulin (Iq) domain herein is meant a region of an immunoglobulin that exists as a distinct structural entity as ascertained by one skilled in the art of protein structure. Ig domains typically have a characteristic ⁇ -sandwich folding topology.
  • immunoglobulin isotype or “isotype” as used herein is meant any of the subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions.
  • the known human immunoglobulin isotypes are lgG1 , lgG2, lgG3, lgG4, lgA1 , lgA2, IgM, IgD, and IgE.
  • IgG as used herein is meant a polypeptide belonging to the class of antibodies that are substantially encoded by a recognized immunoglobulin gamma gene. In humans this class comprises lgG1 , lgG2, lgG3, and lgG4. In mice this class comprises lgG1 , lgG2a, lgG2b, lgG3. As described herein and in the references below, IgG molecules also include hybrid IgG molecules, such as hybrids of lgG1/G2 described in U.S. Publication No.
  • IgG molecules that have amino acid substitutions as compared to wild- type IgG molecules.
  • IgG molecules can include naturally occurring molecules or those with sequence modifications.
  • parent polypeptide By “parent polypeptide”, “parent protein”, “precursor polypeptide”, or “precursor protein” as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant.
  • Said parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide.
  • Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it. Accordingly, by “parent Fc polypeptide” as used herein is meant an Fc polypeptide that is modified to generate a variant, and by “parent antibody” as used herein is meant an antibody that is modified to generate a variant antibody.
  • protein or "polypeptide” as used herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides.
  • the protein may be made up of naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. "analogs", such as peptoids.
  • position as used herein is meant a location in the sequence of a protein or nucleic acid. Protein positions may be numbered sequentially, or according to an established format, for example the Kabat index for antibody variable regions or the EU index for antibody constant regions. For example, position 297 is a position in the human antibody lgG1. Corresponding positions are determined as outlined above, generally through alignment with other parent sequences.
  • residue as used herein is meant a position in a protein and its associated amino acid identity.
  • Asparagine 297 also referred to as Asn297, also referred to as N297
  • Asn297 is a residue in the human antibody lgG1 .
  • it can also refer to nucleic acid bases.
  • target antigen or "target” or “antigen” as used herein is meant the molecule that is bound specifically by the variable region of an antibody, or by the fusion protein of an Fc fusion.
  • a target antigen may be a protein, carbohydrate, lipid, or other chemical compound. A wide variety of suitable targets are described below.
  • target cell as used herein is meant a cell that expresses a target antigen.
  • variant polypeptide polypeptide variant
  • variant polypeptide a polypeptide sequence that differs from that of a parent polypeptide sequence by virtue of at least one amino acid modification.
  • the parent polypeptide may be a naturally occurring or wild-type (WT) polypeptide, or may be a modified version of a WT polypeptide.
  • variant polypeptide may refer to the polypeptide itself, a composition comprising the polypeptide, or the amino sequence that encodes it.
  • the variant polypeptide has at least one amino acid modification compared to the parent polypeptide, e.g. from about one to about ten amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent.
  • the variant polypeptide sequence herein will preferably possess at least about 80% homology with a parent polypeptide sequence, and most preferably at least about 90% homology, more preferably at least about 95% homology. Accordingly, by "Fc variant” or “variant Fc” as used herein is meant an Fc sequence that differs from that of a parent Fc sequence by virtue of at least one amino acid modification.
  • An Fc variant may only encompass an Fc region, or may exist in the context of an antibody, Fc fusion, isolated Fc, Fc fragment, or other polypeptide that is substantially encoded by Fc.
  • Fc variant may refer to the Fc polypeptide itself, compositions comprising the Fc variant polypeptide, or the amino acid sequence that encodes it.
  • Fc polypeptide variant or “variant Fc polypeptide” as used herein is meant an Fc polypeptide that differs from a parent Fc polyeptide by virtue of at least one amino acid modification.
  • Fc variant antibody or “antibody Fc variant” as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification in the Fc region.
  • protein variant or “variant protein” as used herein is meant a protein that differs from a parent protein by virtue of at least one amino acid modification.
  • antibody variant or “variant antibody” as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification.
  • IqG variant or “variant IqG” as used herein is meant an antibody that differs from a parent IgG by virtue of at least one amino acid modification.
  • immunoglobulin variant or “variant immunoqlobluin” as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification.
  • wild type or “WT” or “native” herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations.
  • a WT protein, polypeptide, antibody, immunoglobulin, IgG, etc. has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
  • scFv as used herein is meant a polypeptide consisting of two variable regions connected by a short linker sequence; e.g., V H -linker-VK, V H -linker-VA, VK-linker-V H , or VA- linker-V H .
  • linker sequences consist of but are not limited to (Gly 4 Ser) 3 ,
  • scFv-Fc an scFv fused to a Fc region; e.g., V H -linker-VK-Hinge- CH2-CH3.
  • Antibodies are glycoproteins (although as described herein, engineered glycoforms including aglycosylated and afucosylated forms can be made) having the same structural characteristics. While antibodies exhibit binding specificity to a specific antigen, immunoglobulins include both antibodies and other antibodylike molecules which lack antigen specificity. Polypeptides of the latter kind are, for example, produced at low levels by the lymph system and at increased levels by myelomas.
  • an immunoglobulin is a protein consisting of one or more polypeptides substantially encoded by all or part of at least one immunoglobulin gene. Immunoglobulins may have a number of structural forms, including but not limited to full length antibodies, antibody fragments and analogs (including Fc fusions and immunoadhesins as described below), as well as individual immunoglobulin domains.
  • Antibodies are immunoglobulins that bind a specific antigen. In most mammals, including humans and mice, antibodies are constructed from paired heavy and light polypeptide chains. The light and heavy chain variable regions show significant sequence diversity between antibodies, and are responsible for binding the target antigen. Each chain is made up of individual immunoglobulin (Ig) domains, and thus the generic term
  • immunoglobulin can be used for such proteins.
  • tetramer typically comprises a tetramer. Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one "light” (typically having a molecular weight of about 25 kDa) and one "heavy” chain (typically having a molecular weight of about 50-70 kDa). Human light chains are classified as kappa and lambda light chains. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • IgG has several subclasses, including, but not limited to lgG1 , lgG2, lgG3, and lgG4.
  • IgM has subclasses, including, but not limited to, lgM1 and lgM2.
  • IgA has several subclasses, including but not limited to lgA1 and lgA2.
  • isotype as used herein is meant any of the classes and subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions.
  • the known human immunoglobulin isotypes are lgG1 , lgG2, lgG3, lgG4, lgA1 , lgA2, lgM1 , lgM2, IgD, and IgE.
  • the distinguishing features between these antibody classes are their constant regions, although subtler differences may exist in the variable region.
  • Each of the light and heavy chains are made up of two distinct regions, referred to as the variable and constant regions.
  • the IgG heavy chain is composed of four immunoglobulin domains linked from N- to C-terminus in the order VH-CH1 -CH2-CH3, referring to the heavy chain variable domain, heavy chain constant domain 1 , heavy chain constant domain 2, and heavy chain constant domain 3 respectively (also referred to as VH-Cy1 -Cy2-Cy3, referring to the heavy chain variable domain, constant gamma 1 domain, constant gamma 2 domain, and constant gamma 3 domain respectively).
  • the IgG light chain is composed of two immunoglobulin domains linked from N- to C-terminus in the order VL-CL, referring to the light chain variable domain and the light chain constant domain respectively.
  • the constant regions show less sequence diversity, and are responsible for binding a number of natural proteins to elicit important biochemical events.
  • the structure that constitutes the natural biological form of an antibody, including the variable and constant regions, is referred to herein as a "full length antibody".
  • the full length antibody of the IgG isotype is a tetramer and consists of two identical pairs of two immunoglobulin chains, each pair having one light and one heavy chain, each light chain comprising immunoglobulin domains VL and CL, and each heavy chain comprising immunoglobulin domains VH, Cy1 , Cy2, and Cy3.
  • IgG antibodies may consist of only two heavy chains, each heavy chain comprising a variable domain attached to the Fc region.
  • variable region of an antibody contains the antigen binding determinants of the molecule, and thus determines the specificity of an antibody for its target antigen.
  • the variable region is so named because it is the most distinct in sequence from other antibodies within the same class.
  • the amino-terminal portion of each chain includes a variable region of about 100 to 1 10 or more amino acids primarily responsible for antigen recognition.
  • three loops are gathered for each of the V domains of the heavy chain and light chain to form an antigen-binding site. Each of the loops is referred to as a
  • CDR complementarity-determining region
  • the variable region outside of the CDRs is referred to as the framework (FR) region.
  • FR framework
  • sequence variability does occur in the FR region between different antibodies.
  • this characteristic architecture of antibodies provides a stable scaffold (the FR region) upon which substantial antigen binding diversity (the CDRs) can be explored by the immune system to obtain specificity for a broad array of antigens.
  • a number of high-resolution structures are available for a variety of variable region fragments from different organisms, some unbound and some in complex with antigen.
  • each chain defines a constant region primarily responsible for effector function, e.g. the Fc region.
  • immunoglobulin domains in the heavy chain referred to as heavy constant (CH) regions.
  • the IgG isotypes each have three CH regions. Accordingly, "CH” domains in the context of IgG are as follows: “CH1 " refers to positions 1 18-220 according to the EU index as in Kabat. "CH2" refers to positions 237-340 according to the EU index as in Kabat, and “CH3” refers to positions 341 -447 according to the EU index as in Kabat.
  • hinge region Another important region of the heavy chain is the hinge region.
  • hinge region or “hinge region” or “antibody hinge region” or “immunoglobulin hinge region” herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody. Structurally, the IgG CH1 domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237.
  • the antibody hinge is herein defined to include positions 221 (D221 in lgG1 ) to 236 (G236 in lgG1 ), wherein the numbering is according to the EU index as in Kabat.
  • the lower hinge is included, with the “lower hinge” generally referring to positions 226 or 230 to 236.
  • antibody herein is meant a protein consisting of one or more polypeptides substantially encoded by all or part of the recognized immunoglobulin genes.
  • the recognized immunoglobulin genes include the kappa ( ⁇ ), lambda ( ⁇ ), and heavy chain genetic loci, which together comprise the myriad variable region genes, and the constant region genes mu ( ⁇ ), delta ( ⁇ ), gamma ( ⁇ ), sigma ( ⁇ ), and alpha (a) which encode the IgM, IgD, IgG (lgG1 , lgG2, lgG3, and lgG4), IgE, and IgA (lgA1 and lgA2) isotypes respectively.
  • Antibody herein is meant to include full length antibodies and antibody fragments, and may refer to a natural antibody from any organism, an engineered antibody, or an antibody generated recombinantly for experimental, therapeutic, or other purposes.
  • the Fc region of an antibody interacts with a number of Fc receptors and ligands, imparting an array of important functional capabilities referred to as effector functions.
  • Fc comprises Ig domains Cy2 and Cy3 and the N-terminal hinge leading into Cy2.
  • An important family of Fc receptors for the IgG class are the Fc gamma receptors (FcyRs).
  • this protein family includes FcyRI (CD64), including isoforms FcyRIa, FcyRIb, and FcyRIc; FcyRI I (CD32), including isoforms FcyRlla (including allotypes H 131 and R131 ), FcyRllb (including FcyRl lb- 1 and FcyRl lb-2), and FcyRl lc; and FcyRI II (CD16), including isoforms FcyRl lla (including allotypes V158 and F158) and FcyRll lb (including allotypes FcyRl l lb-NA1 and FcyRl l lb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, hereby entirely incorporated by reference).
  • These receptors typically have an extracellular domain that mediates binding to Fc, a membrane spanning region, and an intracellular domain that may mediate some signaling event within the cell. These receptors are expressed in a variety of immune cells including monocytes, macrophages, neutrophils, dendritic cells, eosinophils, mast cells, platelets, B cells, large granular lymphocytes, Langerhans' cells, natural killer (NK) cells, and ⁇ T cells. Formation of the Fc/FcyR complex recruits these effector cells to sites of bound antigen, typically resulting in signaling events within the cells and important subsequent immune responses such as release of inflammation mediators, B cell activation,
  • ADCC antibody dependent cell-mediated cytotoxicity
  • ADCP antibody dependent cell-mediated phagocytosis
  • the different IgG subclasses have different affinities for the FcyRs, with lgG1 and lgG3 typically binding substantially better to the receptors than lgG2 and lgG4 (Jefferis et al., 2002, Immunol Lett 82:57-65, hereby entirely incorporated by reference).
  • the FcyRs bind the IgG Fc region with different affinities.
  • the extracellular domains of FcyRll la and FcyRl l lb are 96% identical, however FcyRll lb does not have a intracellular signaling domain.
  • FcyRI, FcyRlla/c, and FcyRl lla are positive regulators of immune complex-triggered activation, characterized by having an intracellular domain that has an immunoreceptor tyrosine-based activation motif (ITAM)
  • FcyRl lb has an immunoreceptor tyrosine-based inhibition motif (ITIM) and is therefore inhibitory.
  • activation receptors and FcyRl lb is referred to as an inhibitory receptor.
  • all FcyRs bind the same region on Fc, at the N-terminal end of the Cy2 domain and the preceding hinge.
  • Fc/FcyR binding mediates ADCC
  • Fc/C1 q binding mediates complement dependent cytotoxicity (CDC).
  • a site on Fc between the Cy2 and Cy3 domains mediates interaction with the neonatal receptor FcRn, the binding of which recycles endocytosed antibody from the endosome back to the bloodstream (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12: 181 -220; Ghetie et al., 2000, Annu Rev Immunol 18:739-766, both hereby entirely incorporated by reference).
  • a key feature of the Fc region is the conserved N-linked glycosylation that occurs at N297.
  • This carbohydrate, or oligosaccharide as it is sometimes referred, plays a critical structural and functional role for the antibody, and is one of the principle reasons that antibodies must be produced using mammalian expression systems.
  • Efficient Fc binding to FcyR and C1 q requires this modification, and alterations in the composition of the N297 carbohydrate or its elimination affect binding to these proteins.
  • the antibodies of embodiments disclosed herein may be substantially encoded by immunoglobulin genes belonging to any of the antibody classes.
  • the antibodies disclosed herein comprise sequences belonging to the IgG class of antibodies, including lgG1 , lgG2, lgG3, or lgG4.
  • immunoglobulins disclosed herein comprise sequences belonging to the IgA (including subclasses lgA1 and lgA2), IgD, IgE, IgG, or IgM classes of antibodies.
  • the immunoglobulins disclosed herein may comprise more than one protein chain, e.g., may be an antibody or Fc fusion that is a monomer or an oligomer, including a homo- or hetero-oligomer.
  • Antibodies disclosed herein may be substantially encoded by genes from any organism, e.g., mammals (including, but not limited to humans, rodents (including but not limited to mice and rats), lagomorpha (including but not limited to rabbits and hares), camelidae (including but not limited to camels, llamas, and dromedaries), and non-human primates, including but not limited to Prosimians, Platyrrhini (New World monkeys),
  • the immunoglobulins disclosed herein may be substantially human, with the modifications outlined herein.
  • Gm polymorphism is determined by the IGHG1 , IGHG2 and IGHG3 genes which have alleles encoding allotypic antigenic determinants referred to as G1 m, G2m, and G3m allotypes for markers of the human lgG1 , lgG2 and lgG3 molecules (no Gm allotypes have been found on the gamma 4 chain). Markers may be classified into " allotypes " and
  • isoallotypes are distinguished on different serological bases dependent upon the strong sequence homologies between isotypes. Allotypes are antigenic determinants specified by allelic forms of the Ig genes. Allotypes represent slight differences in the amino acid sequences of heavy or light chains of different individuals. Even a single amino acid difference can give rise to an allotypic determinant, although in many cases there are several amino acid substitutions that have occurred. Allotypes are sequence differences between alleles of a subclass whereby the antisera recognize only the allelic differences.
  • An isoallotype is an allele in one isotype which produces an epitope which is shared with a non- polymorphic homologous region of one or more other isotypes and because of this the antisera will react with both the relevant allotypes and the relevant homologous isotypes (Clark, 1997, IgG effector mechanisms, Chem Immunol. 65:88-1 10; Gorman & Clark, 1990, Semin Immunol 2(6):457-66, both hereby entirely incorporated by reference).
  • G1 m (1 , 2, 3, 17) or G1 m (a, x, f, z), G2m (23) or G2m (n), G3m (5, 6, 10, 1 1 , 13, 14, 15, 16, 21 , 24, 26, 27, 28) or G3m (b1 , c3, b5, bO, b3, b4, s, t, g1 , c5, u, v, g5)
  • the human IgG subclasses molecular analysis of structure, function and regulation. Pergamon, Oxford, pp. 43-78 (1990); Lefranc, G. et al., 1979, Hum.
  • Allotypes that are inherited in fixed combinations are called Gm haplotypes.
  • the immunoglobulins disclosed herein may be substantially encoded by any allotype, isoallotype, or haplotype of any immunoglobulin gene.
  • variable region of an antibody can compose sequences from a variety of species.
  • the antibody variable region can be from a nonhuman source, including but not limited to mice, rats, rabbits, camels, llamas, and monkeys.
  • the scaffold components can be a mixture from different species.
  • an antibody disclosed herein may be a chimeric antibody and/or a humanized antibody.
  • both "chimeric antibodies” and “humanized antibodies” refer to antibodies that combine regions from more than one species.
  • "chimeric antibodies” traditionally comprise variable region(s) from a mouse or other nonhuman species and the constant region(s) from a human.
  • Humanized antibodies generally refer to non-human antibodies that have had the variable-domain framework regions swapped for sequences found in human antibodies.
  • the entire antibody, except the CDRs is encoded by a polynucleotide of human origin or is identical to such an antibody except within its CDRs.
  • the CDRs some or all of which are encoded by nucleic acids originating in a non-human organism, are grafted into the beta-sheet framework of a human antibody variable region to create an antibody, the specificity of which is determined by the engrafted CDRs.
  • the creation of such antibodies is described in, e.g., WO 92/1 1018, Jones, 1986, Nature
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin, and thus will typically comprise a human Fc region.
  • the antibody is a fully human antibody with at least one modification as outlined herein.
  • Fully human antibody or “complete human antibody” refers to a human antibody having the gene sequence of an antibody derived from a human chromosome with the modifications outlined herein. Fully human antibodies may be obtained, for example, using transgenic mice (Lonberg, 2008, Handb Exp Pharmacol 181 :69-97) or human antibody libraries coupled with selection methods (Mondon et al., 2008, Front Biosci 13:1 1 17-29; Lonberg, 2008, Curr Opin Immunol. 20[4]:450-9).
  • antibody analogs may comprise a variety of structures, including, but not limited to full length antibodies, antibody fragments, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as "antibody mimetics"), antibody fusions, antibody
  • the immunogloublin comprises an antibody fragment.
  • Specific antibody fragments include, but are not limited to (i) the Fab fragment consisting of VL, VH, CL and CH1 domains, (ii) the Fd fragment consisting of the VH and CH1 domains, (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment, which consists of a single variable, (v) isolated CDR regions, (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vii) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site, (viii) bispecific single chain Fv dimers, and (ix) "diabodies” or "triabodies", multivalent or multispecific fragments
  • the antibody fragments may be modified.
  • the molecules may be stabilized by the incorporation of disulphide bridges linking the VH and VL domains.
  • disulphide bridges linking the VH and VL domains. Examples of antibody formats and architectures are described in Holliger & Hudson, 2006, Nature Biotechnology 23(9):1 126-1 136, and Carter 2006, Nature Reviews Immunology 6:343-357 and references cited therein, all expressly incorporated by reference.
  • the present invention provides antibody analogs that are a novel set of formats that enable co-engaged antigens to be bound with different valencies. Specifically, as discussed herein, there are two general formats: an antibody analog that has three antigen binding domains, and one that has two.
  • a first antigen can be bound bivalently while a second antigen is simultaneously bound monovalently.
  • the novel immunoglobulin compositions herein co-engage a first and second antigen, referred to herein as antigen-1 and antigen-2 respectively. Binding to the antigens is mediated by first and second antigen binding regions such that antigen-1 is bound bivalently by the immunoglobulin, while antigen-2 is bound monovalently by the immunoglobulin. That is, the immunoglobulin of the invention contains two binding sites for antigen-1 but only one binding site for antigen-2.
  • the antigen binding regions of the immunoglobulin are antibody variable regions, also referred to as Fv regions, each comprising a VH domain and a VL domain.
  • Fv-1 The Fv region that binds antigen-1
  • Fv-2 the Fv region that binds antigen-2
  • Preferred immunoglobulin formats of the invention for co-engaging antigens with different valencies comprise IgG antibodies with a C-terminal Fv region.
  • immunoglobulins typically comprise a full length antibody that is linked C-terminally to a set of VH and VL domains that make up a novel Fv region (Fv-2) and thus a novel antigen binding site (for antigen-2).
  • the C-terminal VH and VL domains may also be linked C- terminally to CH 1 and CL domains, thereby resulting in a C-terminal Fab that makes up the novel antigen binding site.
  • These immunoglobulins are referred to as mAb-Fv and mAb-Fab.
  • the VH and VL domains are typically linked C-terminally to the Fc region via flexible linkers. A variety of linkers are described herein that may find use for linking the VH and VL domains C-terminally to the constant chain in order to generate the novel binding site Fv-2.
  • the N-terminal Fab regions of the mAb-Fv and mAb-Fab contain the binding site for the primary or first antigen (antigen-1 ). Accordingly, this Fv region is referred to as Fv-1 . Because there are two such Fv-1 regions, the mAb-Fv and mAb-Fab bind bivalently to antigen-1 . That is, one immunoglublin binds two of antigen-1 at the same time. In contrast, the C-terminal Fv region of the mAb-Fv or C-terminal Fab region of the mAb-Fab contain the binding site for the second antigen (antigen-2). Thus this Fv region is referred to as Fv-2.
  • the mAb-Fv and mAb-Fab bind monovalently to antigen-2. That is, one immunoglublin binds only one of antigen-2.
  • the mAb-Fv and mAb- Fab immunoglobulins thus contain three variable regions, two of which are Fv-1 and one of which is Fv-2.
  • the mAb-Fv and mAb-Fab immunoglobulins comprise three distinct immunoglobulin chains. These chains include an antibody light chain and two antibody heavy chains. One of said heavy chains comprises a VH or VH-CH 1 at its C-terminus, whereas the other of said heavy chains comprises a VL or VL-CL at its C-terminus.
  • the heavy chain dimer that is competent to bind the second antigen is a heterodimer, rather than a homodimer as is the case naturally for IgG's.
  • the use of two distinct heavy chains is an important component of the mAb-Fv and mAb-Fab that enables the engineering of a second antigen binding site using a VH/VL pair.
  • the mAb-Fv and mAb- Fab formats are particularly suited for the use of variants that favor Fc heterodimerization and disfavor Fc homodimerization.
  • Different variant heavy chains for example containing CH3 substitutions, can be linked to the different heavy chains of the mAb-Fv and mAb-Fab.
  • One variant heavy chain can be linked C-terminally to the VH of the mAb-Fv or VH-CH 1 of the mAb-Fab, while the other variant chain can be linked C-terminally to the VL of the mAb- Fv or VL-CL of the mAb-Fab, thereby generating the new monovalent Fv-2.
  • the additional Fv-2 VH/VL and VH-CH1 /VL-CL pairings may add further specificity to the heterodimer relative to the homodimer.
  • the two 25% homodimer species would comprise homdimeric heavy chains that contain two C-terminal VL's or two C-terminal VH's, and thus would typically not be able to bind antigen-2.
  • the desired 50% heterodimeric mAb-Fv or mAb-Fab species can be purified, yielding a composition that is competent of engaging antigen-2.
  • the antibody analogs of the "trimeric" antigen binding domain constructs generally comprise two modified antibody heavy chains with associated light chains, where each heavy chain possesses two variable regions and a constant region.
  • the first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain- Variable Domain-COOH
  • the second modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Domain-COOH.
  • the N- terminal variable heavy domains interact with the associated light chains to form an antigen binding site on each chain (as in a "traditional” antibody, providing two antigen binding domains).
  • the C-terminus of each chain may further comprise a CH1 domain as well.
  • the first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Domain- Constant Domain (CH1 and/or CL)-COOH
  • the second modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Domain-Constant Domain (CH1 and/or CL)-COOH.
  • Fab-Fv and Fab-Fab are analogues of the mAb-Fv and mAb-Fab that bind both antigen-1 and antigen-2 monovalently. These immunoglobulins are referred to as Fab-Fv and Fab-Fab. Rather than comprising three separate chains, these formats contain two different heavy chains, i.e. they lack a native light chain. For one of the heavy chains, the N- terminal Fv-1 VH-CH1 half of the Fab region are replaced with the Fv-1 VL-CL region.
  • the heterodimer species of the Fab-Fv and Fab-Fab comprise an N-terminal Fab region that contains a single binding site for antigen-1 , and comprise a C-terminal Fv or Fab region that contains a single binding site for antigen-2.
  • the first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Heavy Domain-COOH
  • the second modified antibody heavy chain has the structure NH2- Variable Light Domain-Constant Domain-Variable Light Domain.
  • the first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Heavy Domain-Constant Domain (e.g., CH1 )-COOH
  • the second modified antibody heavy chain has the structure NH2-Variable Light Domain-Constant Domain-Variable Light Domain-Constant Domain (e.g., CL)-COOH.
  • VHH regions are described, for example, in Wesolowski et al., 2009, Med Microbiol Immunol 198[3]:157-174, and references therein, herein expressly incorporated by reference.
  • the N-terminal binding portion of the mAb-Fv and mAb-Fab may lack a light chain and a CH1 domain. It is also possible to utilize the C-terminal monovalent Fv-2 in the context of an Fc fusion, also referred to as an immunoadhesin. In this format, Fv- 1 is replaced with a fusion protein, for example a receptor that binds antigen-1. Thus antigen-1 is bound by a fusion protein, while antigen-2 is bound by Fv-2.
  • Antibody analogs disclosed herein may comprise an Fc variant.
  • An Fc variant comprises one or more amino acid modifications relative to a parent Fc polypeptide, wherein the amino acid modification(s) provide one or more optimized properties.
  • modification herein is meant an alteration in the physical, chemical, or sequence properties of a protein, polypeptide, antibody, or immunoglobulin.
  • An amino acid modification can be an amino acid substitution, insertion, and/or deletion in a polypeptide sequence.
  • amino acid substitution or “substitution” herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with another amino acid.
  • substitution Y349T refers to a variant polypeptide, in this case a constant heavy chain variant, in which the tyrosine at position 349 is replaced with threonine.
  • amino acid insertion or “insertion” as used herein is meant the addition of an amino acid at a particular position in a parent polypeptide sequence.
  • amino acid deletion or “deletion” as used herein is meant the removal of an amino acid at a particular position in a parent polypeptide sequence.
  • An Fc variant disclosed herein differs in amino acid sequence from its parent by virtue of at least one amino acid modification.
  • the immunoglobulins disclosed herein may have more than one amino acid modification as compared to the parent, for example from about one to fifty amino acid modifications, e.g., from about one to ten amino acid
  • the sequences of the Fc variants and those of the parent Fc polypeptide are substantially homologous.
  • the variant Fc variant sequences herein will possess about 80% homology with the parent Fc variant sequence, e.g., at least about 90% homology, at least about 95% homology, at least about 98% homology, at least about 99% homology, etc.
  • Modifications disclosed herein also include glycoform modifications.
  • Modifications may be made genetically using molecular biology, or may be made
  • Fc variants disclosed herein are defined according to the amino acid modifications that compose them.
  • substitution Y349T refers to a variant
  • polypeptide in this case a constant heavy chain variant, in which the tyrosine at position 349 is replaced with threonine.
  • Y349T/T394F defines an Fc variant with the
  • substitutions Y349T and T394F relative to the parent Fc polypeptide may be unspecified, in which case the aforementioned variant is referred to as 349T/394F. It is noted that the order in which substitutions are provided is arbitrary, that is to say that, for example, 349T/394F is the same Fc variant as 394F/349T. Unless otherwise noted, constant region and Fc positions discussed herein are numbered according to the EU index or EU numbering scheme (Kabat et al., 1991 , Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health,
  • the Fc variants disclosed herein are based on human IgG sequences, and thus human IgG sequences are used as the "base" sequences against which other sequences are compared, including but not limited to sequences from other organisms, for example rodent and primate sequences. Immunoglobulins may also comprise sequences from other immunoglobulin classes such as IgA, IgE, IgGD, IgGM, and the like.
  • the variants disclosed herein are engineered in the context of one parent IgG, the variants may be engineered in or "transferred” to the context of another, second parent IgG. This is done by determining the "equivalent” or “corresponding” residues and substitutions between the first and second IgG, typically based on sequence or structural homology between the sequences of the first and second IgGs. In order to establish homology, the amino acid sequence of a first IgG outlined herein is directly compared to the sequence of a second IgG.
  • the residues equivalent to particular amino acids in the primary sequence of the first immunoglobulin are defined. Alignment of conserved residues may conserve 100% of such residues. However, alignment of greater than 75% or as little as 50% of conserved residues is also adequate to define equivalent residues. Equivalent residues may also be defined by determining structural homology between a first and second IgG that is at the level of tertiary structure for IgGs whose structures have been determined.
  • equivalent residues are defined as those for which the atomic coordinates of two or more of the main chain atoms of a particular amino acid residue of the parent or precursor (N on N, CA on CA, C on C and O on O) are within about 0.13 nm, after alignment. In another embodiment, equivalent residues are within about 0.1 nm after alignment. Alignment is achieved after the best model has been oriented and positioned to give the maximum overlap of atomic coordinates of non-hydrogen protein atoms of the proteins.
  • the Fc variants discovered as disclosed herein may be engineered into any second parent IgG that has significant sequence or structural homology with the Fc variant.
  • the variant antibody may be engineered in another lgG1 parent antibody that binds a different antigen, a human lgG2 parent antibody, a human IgA parent antibody, a mouse lgG2a or lgG2b parent antibody, and the like.
  • the context of the parent Fc variant does not affect the ability to transfer the Fc variants disclosed herein to other parent IgGs.
  • Fc variants that favor heterodimerization of the Fc region relative to homodimerization. That is, by modifying one or more amino acids within each heavy chain that forms the Fc region, such that each heavy chain comprises at least one amino acid variant as compared to the other chain, binding of the different heavy chains to form heterodimeric Fc regions is favored over the individual heavy chains forming homodimers.
  • hetero-Fc variants comprise a pair of different variants such that one pair (referred to as variant') modifies one CH3 domain (referred to as CH3'), while the other pair (variant”) modifies another CH3 domain (CH3").
  • variants are designed such that their interaction in a heterodimeric CH3 interface (variant'/variant”) is favorable, thus resulting in a stable heterodimeric Fc species (comprising CH37CH3").
  • variant'/variant' and variant'Vvariant is unfavorable, resulting in unstable homodimeric Fc species (comprising CH3VCH3' and CH37CH3").
  • hetero-Fc variants of the invention increase the percentage population of the heterodimer while reducing the population of the homodimers.
  • Fc variants that favor heterodimerization and disfavor homodimerization may alter the populations such that the heterodimer population increases to 70%, preferably 80%, more preferably 90%, and most preferably greater than 95%.
  • hetero-Fc variants may reduce the homodimer populations to 2.5% and 2.5% while increasing the heterodimer population to 95%.
  • Populations of the different species may be measured using a variety of methods well-known in the art, including methods described herein, including for example gel electropheresis and biochemical assays.
  • Heterodimeric immunogloublin species may optionally be isolated further from homodimeric species using any number of various protein purification methods well-known in the art.
  • Hetero-Fc variants herein preferably comprise at least one substitution at a position in a CH3 domain selected from the group consisting of 349, 351 , 354, 356, 357, 364, 366, 368, 370, 392, 394, 395, 396, 397, 399, 401 , 405, 407, 409, 41 1 , and 439, wherein numbering is according to the EU index as in Kabat.
  • hetero-Fc variants comprise at least one CH3 domain substitution per heavy chain selected from the group consisting of 349A, 349C, 349E, 349I, 349K, 349S, 349T, 349W, 351 E, 351 K, 354C, 356K, 357K, 364C, 364D, 364E, 364F, 364G, 364H, 364R, 364T, 364Y, 366D, 366K, 366S, 366W, 366Y, 368A, 368E, 368K, 368S, 370C, 370D, 370E, 370G, 370R, 370S, 370V, 392D, 392E, 394F, 394S, 394W, 394Y, 395T, 395V, 396T, 397E, 397S, 397T, 399K, 401 K, 405A, 405S, 407T, 407V, 4
  • each heavy chain can comprise different numbers of subsitutions. As shown in Tables 2 and 3, below, both heavy chains that make up the Fc region may comprise a single substitution, one chain may comprise a single substitution and the other two substitutions, both can contain two substitutions (although each chain will contain different substitutions), etc.
  • the hetero-Fc variants are made in combinations, that is, two or more variants per heavy chain Fc domain, selected from the group outlined above.
  • combination variants including, but not limited to, those shown in Tables 2 and 3 and in the Figures.
  • Fc variants disclosed herein may be optimized for improved or reduced binding to Fc receptors or Fc ligands.
  • Fc receptor or “Fc ligand” as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc-ligand complex.
  • Fc ligands include but are not limited to FcyRs, (as described above, including but not limited to FcyRllla, FcyRlla, FcyRllb, FcyRI and FcRn), C1 q, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral FcyR.
  • Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the FcyRs.
  • Fc ligands may include
  • Immunoglobulins herein may be engineered to for optimized properties, including but are not limited to enhanced or reduced affinity for an Fc receptor.
  • greater affinity or “improved affinity” or “enhanced affinity” or “better affinity” than a parent Fc polypeptide
  • an Fc variant binds to an Fc receptor with a significantly higher equilibrium constant of association (KA or K a ) or lower equilibrium constant of dissociation (KD or K d ) than the parent Fc polypeptide when the amounts of variant and parent polypeptide in the binding assay are essentially the same.
  • the Fc variant with improved Fc receptor binding affinity may display from about 5 fold to about 1000 fold, e.g.
  • Fc receptor binding affinity is determined, for example, by the binding methods disclosed herein, including but not limited to Biacore, by one skilled in the art. Accordingly, by “reduced affinity" as compared to a parent Fc polypeptide as used herein is meant that an Fc variant binds an Fc receptor with significantly lower KA or higher KD than the parent Fc polypeptide. Greater or reduced affinity can also be defined relative to an absolute level of affinity.
  • particularly useful Fc modifications for the present invention are variants that reduce or ablate binding to one or more FcyRs and/or complement proteins, thereby reducing or ablating Fc-mediated effector functions such as ADCC, ADCP, and CDC.
  • Such variants are also referred to herein as “knockout variants” or "KO variants”.
  • Variants that reduce binding to FcyRs and complement are useful for reducing unwanted interactions mediated by the Fc region and for tuning the selectivity of the immunoglobulins.
  • Preferred knockout variants are described in USSN1 1/981 ,606, filed 10/31/2007, entitled “Fc Variants with Optimized Properties," herein expressly incorporated by reference.
  • Preferred modifications include but are not limited substitutions, insertions, and deletions at positions 234, 235, 236, 237, 267, 269, 325, and 328, wherein numbering is according to the EU index.
  • substitutions include but are not limited to 234G, 235G, 236R, 237K, 267R, 269R, 325L, and 328R, wherein numbering is according to the EU index.
  • a preferred variant comprises 236R/328R.
  • Variants may be used in the context of any IgG isotype or IgG isotype Fc region, including but not limited to human lgG1 , lgG2, lgG3, and/or lgG4 and combinations thereof.
  • Preferred IgG Fc regions for reducing FcyR and complement binding and reducing Fc-mediated effector functions are lgG2 and lgG4 Fc regions.
  • Hybrid isotypes may also be useful, for example hybrid lgG1/lgG2 isotypes as described in
  • Fc modifications that improve binding to FcyRs and/or complement may also find use in the immunoglobulins herein. Such Fc variants may enhance Fc-mediated effector functions such as ADCC, ADCP, and/or CDC. Preferred modifications for improving FcyR and complement binding are described in USSN1 1/124,620 (with Figure 41 being expressly incorporated by reference in its entirety for the modifications depicted therein) and
  • Preferred modifications comprise a substitution at a position selected from the group consisting of 236, 239, 268, 324, and 332, wherein numbering is according to the EU index.
  • Preferred substitutions include but are not limited to 236A, 239D, 239E, 268D, 267E, 268E, 268F, 324T, 332D, and 332E.
  • Preferred variants include but are not limited to 239D/332E, 236A/332E,
  • the immunoglobulins disclosed herein may incorporate Fc variants that enhance affinity for an inhibitory receptor FcyRllb. Such variants may provide the immunoglobulins herein with immunomodulatory activities related to FcyRllb + cells, including for example B cells and monocytes. In one embodiment, the Fc variants provide selectively enhanced affinity to FcyRllb relative to one or more activating receptors.
  • Fc variants that improve binding to FcyRllb may include one or more modifications at a position selected from the group consisting of 234, 235, 236, 237, 239, 266, 267, 268, 325, 326, 327, 328, and 332, according to the EU index.
  • substitutions for enhancing FcyRllb affinity include but are not limited to 234D, 234E, 234W, 235D, 235F, 235R, 235Y, 236D, 236N, 237D, 237N, 239D, 239E, 266M, 267D, 267E, 268D, 268E, 327D, 327E, 328F, 328W, 328Y, and 332E. More preferably, substitutions include but are not Imited to 235Y, 236D, 239D, 266M, 267E, 268D, 268E, 328F, 328W, and 328Y.
  • Preferred Fc variants for enhancing binding to FcyRllb include but are not limited to 235Y/267E, 236D/267E, 239D/268D, 239D/267E, 267E/268D, 267E/268E, and 267E/328F.
  • immunoglobulins disclosed herein may incorporate Fc variants that improve FcRn binding. Such variants may enhance the in vivo pharmacokinetic properties of the immunoglobulins.
  • Preferred variants that increase binding to FcRn and/or improve pharmacokinetic properties include but are not limited to substitutions at positions 259, 308, 428, and 434, including but not limited to for example 259I, 308F, 428L, 428M, 434S, 434H, 434F, 434Y, 434M, 428L/434S, 259I/308F and 259I/308F/428L (and others described in USSN12/341.769, filed Dec.
  • Fc Variants with Altered Binding to FcRn include but are not limited to: 250E, 250Q, 428L, 428F, 250Q/428L (Hinton et al., 2004, J. Biol. Chem. 279(8): 6213-6216, Hinton et al.
  • Immunoglobulins described herein can incorporate Fc modifications in the context of any IgG isotype or IgG isotype Fc region, including but not limited to human lgG1 , lgG2, lgG3, and/or lgG4.
  • the IgG isotype may be selected such as to alter FcyR- and/or complement- mediated effector fu notion (s).
  • Hybrid IgG isotypes may also be useful.
  • USSN1 1/256,060 describes a number of hybrid lgG1/lgG2 constant regions that may find use in the particular invention.
  • the invention
  • immunoglobulins may comprise means for isotypic modifications, that is, modifications in a parent IgG to the amino acid type in an alternate IgG.
  • an lgG1/lgG3 hybrid variant may be constructed by a substitutional means for substituting lgG1 positions in the CH2 and/or CH3 region with the amino acids from lgG3 at positions where the two isotypes differ.
  • a hybrid variant IgG antibody may be constructed that comprises one or more substitutional means, e.g., 274Q, 276K, 300F, 339T, 356E, 358M, 384S, 392N, 397M, 422I, 435R, and 436F.
  • an lgG1/lgG2 hybrid variant may be constructed by a substitutional means for substituting lgG2 positions in the CH2 and/or CH3 region with amino acids from lgG1 at positions where the two isotypes differ.
  • a hybrid variant IgG antibody may be constructed that comprises one or more substitutional means, e.g., one or more of the following amino acid substations: 233E, 234L, 235L, -236G
  • the immunoglobulins of the invention may target virtually any antigen or pair of antigens.
  • the mAb-Fv and mAb-Fab are particularly beneficial for targeting distinct antigens, that is where antigen-1 and antigen-2 are different.
  • the immunoglobulins herein preferably co-engage two target antigens.
  • Particular suitable applications of the immunoglobulins herein are co-target pairs for which it is beneficial or critical to engage a target antigen monovalently.
  • antigens may be, for example, immune receptors that are activated upon immune complexation. Cellular activation of many immune receptors occurs only by cross-linking, acheived typically by antibody/antigen immune complexes, or via effector cell to target cell engagement.
  • activation only upon engagement with co-engaged target is critical, as nonspecifiic cross-linking in a clinical setting can elicit a cytokine storm and toxicity.
  • the ability to target two different antigens with different valencies is a novel and useful aspect of the present invention.
  • target antigens for which it may be therapeutically beneficial or necessary to co-engage monovalently include but are not limited to immune activating receptors such as CD3, FcyRs, toll-like receptors (TLRs) such as TLR4 and TLR9, cytokine, chemokine, cytokine receptors, and chemokine receptors.
  • immune activating receptors such as CD3, FcyRs, toll-like receptors (TLRs) such as TLR4 and TLR9
  • cytokine chemokine
  • cytokine receptors chemokine receptors
  • chemokine receptors chemokine receptors
  • mAb-Fv and mAb-Fab immunoglobulins described herein are particularly beneficial for targeting distinct antigens, in some cases it may be beneficial to target only one antigen. That is, Fv-1 and Fv-2 may have specificity for the same antigen, thus binding the target antigen trivalently.
  • any antigen may be targeted by the immunoglobulins herein, including but not limited to proteins, subunits, domains, motifs, and/or epitopes belonging to the following list of target antigens, which includes both soluble factors such as cytokines and membrane- bound factors, including transmembrane receptors: 17-IA, 4-1 BB, 4Dc, 6-keto-PGF1 a, 8-iso- PGF2a, 8-oxo-dG, A1 Adenosine Receptor, A33, ACE, ACE-2, Activin, Activin A, Activin AB, Activin B, Activin C, Activin RIA, Activin RIA ALK-2, Activin RIB ALK-4, Activin RIIA, Activin RU B, ADAM, ADAM10, ADAM 12, ADAM 15, ADAM17/TACE, ADAM8, ADAM9, ADAMTS, ADAMTS4, ADAMTS5, Address
  • RANTES RANTES
  • Relaxin A-chain Relaxin B-chain
  • renin respiratory syncytial virus (RSV) F
  • RSV respiratory syncytial virus
  • RPA2 RSK
  • S100 SCF/KL
  • SDF-1 SDF-1
  • SERINE Serum albumin
  • sFRP-3 Shh, SIGIRR, SK-1 , SLAM, SLPI, SMAC, SMDF, SMOH, SOD, SPARC, Stat, STEAP, STEAP-II, TACE, TACI, TAG-72 (tumor-associated
  • T-cell receptors e.g., T-cell receptor alpha/beta
  • TdT TECK
  • TEM1 TEM5
  • TEM5 TEM5
  • TEM7 TEM7
  • TEM8 testicular PLAP-like alkaline phosphatase
  • TfR TGF
  • TGF-alpha TGF-beta
  • TGF-beta Pan Specific, TGF-beta Rl (ALK-5), TGF-beta Rll, TGF-beta Rllb, TGF-beta Rill, TGF-beta1 , TGF-beta2, TGF-beta3, TGF-beta4, TGF-beta5, Thrombin, Thymus Ck-1 , Thyroid stimulating hormone, Tie, TIMP, TIQ, Tissue Factor, TMEFF2, Tmpo, TMPRSS2, TNF, TNF-alpha, TNF-alpha beta, TNF-beta2, TNFc,
  • TNFRSF14 HVEM ATAR, HveA, LIGHT R, TR2
  • TNFRSF16 NGFR p75NTR
  • TNFRSF17 BCMA
  • TNFRSF18 GITR AITR
  • TNFRSF19 TROY TAJ, TRADE
  • TNFRSF19L RELT
  • TNFRSF1A (TNF Rl CD120a, p55-60)
  • TNFRSF1 B (TNF Rl l CD 120b, p75-80)
  • TNFRSF26 TNFRH3
  • TNFRSF3 LbR TNF Ri ll, TNFC R
  • TNFRSF4 OF40 ACT35, TXGP1 R
  • TNFRSF5 CD40 p50
  • TNFRSF6 Feas Apo-1 , APT1 , CD95
  • TN FRSF6B DcR3 M68, TR6
  • TNFRSF7 (CD27)
  • TNFRSF8 (CD30)
  • Exemplary antigens that may be targeted specifically by the immunoglobulins of the invention include but are not limited to: CD20, CD19, Her2, EGFR, EpCAM, CD3, FcyRl lla (CD16), FcyRl la (CD32a), FcyRl lb (CD32b), FcyRI (CD64), Toll-like receptors (TLRs) such as TLR4 and TLR9, cytokines such as I L-2, IL-5, I L-13, I L-12, IL-23, and TNFa, cytokine receptors such as I L-2R, chemokines, chemokine receptors, growth factors such as VEGF and HGF, and the like.
  • TLRs Toll-like receptors
  • cytokines such as I L-2, IL-5, I L-13, I L-12, IL-23, and TNFa
  • cytokine receptors such as I L-2R
  • chemokines chemokine receptors
  • target antigens and co-targets depend on the desired therapeutic application. Some targets that have proven especially amenable to antibody therapy are those with signaling functions. Other therapeutic antibodies exert their effects by blocking signaling of the receptor by inhibiting the binding between a receptor and its cognate ligand. Another mechanism of action of therapeutic antibodies is to cause receptor down regulation. Other antibodies do not work by signaling through their target antigen.
  • the choice of co-targets will depend on the detailed biology underlying the pathology of the indication that is being treated.
  • co-targets for oncology include but are not limited to HGF and VEGF, IGF-1 R and VEGF, Her2 and VEGF, CD19 and CD3, CD20 and CD3, Her2 and CD3, CD19 and FcyRN Ia, CD20 and FcyRN Ia, Her2 and FcyRNIa.
  • An immunoglobulin of the invention may be capable of binding VEGF and phosphatidylserine; VEGF and ErbB3; VEGF and PLGF; VEGF and ROB04; VEGF and BSG2; VEGF and CDCP1 ; VEGF and ANPEP; VEGF and c-MET; HER-2 and ERB3; HER-2 and BSG2; HER-2 and CDCP1 ; H ER-2 and ANPEP; EGFR and CD64; EGFR and BSG2; EGFR and CDCP1 ; EGFR and ANPEP; IGF1 R and PDGFR; IGF1 R and VEGF; IGF1 R and CD20; CD20 and CD74; CD20 and CD30; CD20 and DR4; CD20 and VEGFR2; CD20 and CD52; CD20 and CD4; HGF and c-MET; HGF and NRP1 ; HGF and phosphatidylserine; ErbB3 and IGF1
  • immunoglobulins herein may bind include, but are not limited to those selected from the group consisting of: CD52, CD20, CD19, CD3, CD4, CD8, BMP6, IL12A, IL1A, IL1B, 1L2, IL24, INHA, TNF, TNFSF10, BMP6, EGF, FGF1, FGF10, FGF11, FGF12, FGF13, FGF14, FGF16, FGF17, FGF18, FGF19, FGF2, FGF20, FGF21, FGF22, FGF23, FGF3, FGF4, FGF5, FGF6, FGF7, FGF8, FGF9, GRP, IGF1, IGF2, IL12A, IL1A, IL1B, 1L2, INHA, TGFA, TGFB1, TGFB2, TGFB3, VEGF, CDK2, FGF10, FGF18, FGF2, FGF4, FGF7, IGF1R, IL2, BCL2, CD164, CDKN1A
  • Autoimmune and inflammatory targets include but are not limited to C5, CCL1 (I-309), CCL1 1 (eotaxin), CCL13 (mcp-4), CCL15 (MIP-1 d), CCL16 (HCC-4), CCL17 (TARC), CCL18 (PARC), CCL19, CCL2 (mcp-1 ), CCL20 (MIP-3a), CCL21 (MIP-2), CCL23 (MPIF-1 ), CCL24 (MPIF-2/eotaxin-2), CCL25 (TECK), CCL26, CCL3 (MIP-1 a), CCL4 (MIP- 1 b), CCL5 (RANTES), CCL7 (mcp-3), CCL8 (mcp-2), CXCL1 , CXCL10 (IP-10), CXCL1 1 (1 - TAC/IP-9), CXCL12 (SDF1 ), CXCL13, CXCL14, CXCL2, CXCL3, CXCL5 (ENA-78/LIX
  • TNFSF5 TNFSF6, TNFSF11, VEGF, ZFPM2, and RNF110 (ZNF144).
  • Exemplary co-targets for autoimmune and inflammatory disorders include but are not limited to IL-1 and TNFalpha, IL-6 and TNFalpha, IL-6 and IL-1, IgE and IL-13, IL-1 and IL- 13, IL-4 and IL-13, IL-5 and IL-13, IL-9 and IL-13, CD19 and FcyRllb, and CD79 and
  • Immunglobulins of the invention with specificity for the following pairs of targets to treat inflammatory disease are contemplated: TNF and IL-17A; TNF and RANKL; TNF and VEGF; TNF and SOST; TNF and DKK; TNF and alphaVbeta3; TNF and NGF; TNF and IL- 23p19; TNF and IL-6; TNF and SOST; TNF and IL-6R; TNF and CD-20; IgE and IL-13; IL-13 and IL23p19; IgE and IL-4; IgE and IL-9; IgE and IL-9; IgE and IL-13; IL-13 and IL-9; IL-13 and IL-4; IL-13 and IL-9; IL-13 and IL-9; IL-13 and IL-4; IL-13 and IL-9; IL-13 and IL-9; IL-13 and IL-4; IL-13 and IL-23p19; IL-13 and IL-9; IL-6R and
  • targets that the immunoglobulins described herein can bind and be useful to treat asthma may be determined.
  • targets include, but are not limited to, IL-13 and IL-1 beta, since IL-1 beta is also implicated in inflammatory response in asthma; IL-13 and cytokines and chemokines that are involved in inflammation, such as IL-13 and IL- 9; IL-13 and IL-4; IL-13 and IL-5; IL-13 and IL-25; IL-13 and TARC; IL-13 and MDC; IL-13 and MIF; IL-13 and TGF- ⁇ ; IL-13 and LHR agonist; IL-13 and CL25; IL-13 and SPRR2a; IL- 13 and SPRR2b; and IL-13 and ADAM8.
  • the immunoglobulins herein may have specifity for one or more targets involved in asthma selected from the group consisting of CSF1 (MCSF), CSF2 (GM-CSF), CSF3 (GCSF), FGF2, IFNA1 , IFNB1 , IFNG, histamine and histamine receptors, IL1A, IL1 B, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL1 1 , IL12A, IL12B, IL13, IL14, IL15, IL16, IL17, IL18, IL19, KITLG, PDGFB, IL2RA, IL4R, IL5RA, IL8RA, IL8RB, IL12RB1 , IL12RB2, IL13RA1 , IL13RA2, IL18R1 , TSLP, CCLi, CCL2, CCL3, CCL4, CCL5, CCL7, CCL
  • Pairs of targets involved in rheumatoid arthritis may be co-targeted by the invention, including but not limited to TNF and IL-18; TNF and IL-12; TNF and IL-23; TNF and IL-1 beta; TNF and MIF; TNF and IL-17; and TNF and IL-15.
  • Antigens that may be targeted in order to treat systemic lupus erythematosus (SLE) by the immunoglobulins herein include but are not limited to CD-20, CD-22, CD-19, CD28, CD4, CD80, HLA-DRA, IL10, IL2, IL4, TNFRSF5, TNFRSF6, TNFSF5, TNFSF6, BLR1 , HDAC4, HDAC5, HDAC7A, HDAC9, ICOSL, IGBP1 , MS4A1 , RGSI, SLA2, CD81 , IFNB1 , IL10, TNFRSF5, TNFRSF7, TNFSF5, AICDA, BLNK, GALNAC4S-6ST, HDAC4, HDAC5, HDAC7A, HDAC9, IL10, IL1 1 , IL4, INHA, INHBA, KLF6, TNFRSF7, CD28, CD38, CD69, CD80, CD83, CD86, DPP4, FCER2,
  • the immunoglobulins herein may target antigens for the treatment of multiple sclerosis (MS), inlcuding but not limited to IL-12, TWEAK, IL-23, CXCL13, CD40, CD40L, IL- 18, VEGF, VLA-4, TNF, CD45RB, CD200, IFNgamma, GM-CSF, FGF, C5, CD52, and CCR2.
  • MS multiple sclerosis
  • An embodiment includes co-egagement of anti-IL-12 and TWEAK for the treatment of MS.
  • One aspect of the invention pertains to immunoglobulins capable of binding one or more targets involved in sepsis, in an embodiment two targets, selected from the group consisting TNF, IL-1 , MIF, IL-6, IL-8, IL-18, IL-12, IL-23, FasL, LPS, Toll-like receptors, TLR- 4, tissue factor, MIP-2, ADORA2A, CASP1 , CASP4, IL-10, IL-1 B, N FKB1 , PROC,
  • immunoglobulins herein may be directed against antigens for the treatment of infectious diseases.
  • any of the aforementioned target antigens, the ligands or receptors that bind them, or other members of their corresponding biochemical pathway may be operably linked to the immunoglobulins of the present invention in order to generate an Fc fusion.
  • an Fc fusion that targets TNFa could be constructed by operably linking an Fc region to TNFR1 or TNFR2.
  • biochemical pathways may be operably linked to the immunoglobulins of the present invention to develop an Fc fusion.
  • linker is used to denote polypeptides comprising two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions.
  • linker polypeptides are well known in the art (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1 121-1 123).
  • a variety of linkers may find use in some embodiments described herein to covalently link Fc regions to a fusion partner.
  • Linker herein is also referred to as "linker sequence”, “spacer”, “tethering sequence” or grammatical equivalents thereof. Homo-or hetero-bifunctional linkers as are well known (see, 1994 Pierce Chemical Company catalog, technical section on cross- linkers, pages 155-200, incorporated entirely by reference). A number of strategies may be used to covalently link molecules together. These include, but are not limited to polypeptide linkages between N- and C-termini of proteins or protein domains, linkage via disulfide bonds, and linkage via chemical cross-linking reagents. In one aspect of this embodiment, the linker is a peptide bond, generated by recombinant techniques or peptide synthesis.
  • the linker peptide may predominantly include the following amino acid residues: Gly, Ser, Ala, or Thr.
  • the linker peptide should have a length that is adequate to link two molecules in such a way that they assume the correct conformation relative to one another so that they retain the desired activity.
  • the linker is from about 1 to 50 amino acids in length, preferably about 1 to 30 amino acids in length. In one embodiment, linkers of 1 to 20 amino acids in length may be used.
  • Useful linkers include glycine-serine polymers, including for example (GS)n, (GSGGS)n, (GGGGS)n, and (GGGS)n, where n is an integer of at least one, glycine-alanine polymers, alanine-serine polymers, and other flexible linkers.
  • nonproteinaceous polymers including but not limited to
  • polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol may find use as linkers, that is may find use as linkers.
  • linker sequences may include any sequence of any length of CL/CH 1 domain but not all residues of CL/CH 1 domain; for example the first 5-12 amino acid residues of the CL/CH 1 domains.
  • Linkers can be derived from immunoglobulin light chain, for example CK or C .
  • Linkers can be derived from immunoglobulin heavy chains of any isotype, including for example Cy1 , Cy2, Cy3, Cy4, Ca1 , Ca2, C8, Cs, and ⁇ .
  • Linker sequences may also be derived from other proteins such as Ig-like proteins (e.g. TCR, FcR, KI R), hinge region- derived sequences, and other natural sequences from other proteins.
  • All antibodies contain carbohydrate at conserved positions in the constant regions of the heavy chain. Each antibody isotype has a distinct variety of N-linked carbohydrate structures. Aside from the carbohydrate attached to the heavy chain, up to 30% of human IgGs have a glycosylated Fab region. IgG has a single N-linked biantennary carbohydrate at Asn297 of the CH2 domain. For IgG from either serum or produced ex vivo in hybridomas or engineered cells, the IgG are heterogeneous with respect to the Asn297 linked
  • the core oligosaccharide normally consists of
  • glycosylation means the attachment of oligosaccharides (carbohydrates containing two or more simple sugars linked together e.g. from two to about twelve simple sugars linked together) to a glycoprotein.
  • oligosaccharide side chains are typically linked to the backbone of the glycoprotein through either N- or O-linkages.
  • the oligosaccharides of immunoglobulins disclosed herein occur generally are attached to a CH2 domain of an Fc region as N-linked oligosaccharides.
  • N-linked glycosylation refers to the attachment of the carbohydrate moiety to an asparagine residue in a glycoprotein chain.
  • each of murine lgG1 , lgG2a, lgG2b and lgG3 as well as human lgG1 , lgG2, lgG3, lgG4, IgA and IgD CH2 domains have a single site for N-linked
  • a "mature core carbohydrate structure” refers to a processed core carbohydrate structure attached to an Fc region which generally consists of the following carbohydrate structure GlcNAc(Fucose)-GlcNAc-Man-(Man-GlcNAc)2 typical of biantennary oligosaccharides.
  • the mature core carbohydrate structure is attached to the Fc region of the glycoprotein, generally via N-linkage to Asn297 of a CH2 domain of the Fc region.
  • a "bisecting GlcNAc” is a GlcNAc residue attached to the a1 ,4 mannose of the mature core carbohydrate structure.
  • the bisecting GlcNAc can be enzymatically attached to the mature core carbohydrate structure by a a(1 ,4)-N-acetylglucosaminyltransferase III enzyme (GnTIII).
  • GnTIII a(1 ,4)-N-acetylglucosaminyltransferase III enzyme
  • CHO cells do not normally express GnTIII (Stanley et al., 1984, J. Biol. Chem. 261 :13370-13378), but may be engineered to do so (Umana et al., 1999, Nature Biotech. 17:176-180).
  • modified glycoforms or engineered glycoforms as used herein is meant a carbohydrate composition that is covalently attached to a protein, for example an antibody, wherein said carbohydrate composition differs chemically from that of a parent protein.
  • Engineered glycoforms may be useful for a variety of purposes, including but not limited to enhancing or reducing FcyR-mediated effector function.
  • the immunoglobulins disclosed herein are modified to control the level of fucosylated and/or bisecting oligosaccharides that are covalently attached to the Fc region.
  • oligosaccharides that are covalently attached to the Fc region, for example by expressing an IgG in various organisms or cell lines, engineered or otherwise (for example Lec-13 CHO cells or rat hybridoma YB2/0 cells), by regulating enzymes involved in the glycosylation pathway (for example FUT8 [a1 ,6-fucosyltranserase] and/or ⁇ 1-4- N- acetylglucosaminyltransferase III [GnTIII]), by modifying carbohydrate(s) after the IgG has been expressed, or by expressing antibody in the presence of fucose analogs as enzymatic inhibitors.
  • Other methods for modifying glycoforms of the immunoglobulins disclosed herein include using glycoengineered strains of yeast (Li et al., 2006, Nature Biotechnology
  • immunoglobulins disclosed herein are glycoengineered to alter the level of sialylation.
  • Higher levels of sialylated Fc glycans in immunoglobulin G molecules can adversely impact functionality (Scallon et al., 2007, Mol Immunol. 44(7):1524-34), and differences in levels of Fc sialylation can result in modified anti-inflammatory activity (Kaneko et al., 2006, Science 313:670-673). Because antibodies may acquire anti-inflammatory properties upon sialylation of Fc core polysaccharide, it may be advantageous to
  • Engineered glycoform typically refers to the different carbohydrate or
  • an immuoglobulin may comprise an engineered glycoform.
  • engineered glycoform may refer to the immunoglobulin that comprises the different carbohydrate or oligosaccharide.
  • a composition disclosed herein comprises a glycosylated immunoglobulin having an Fc region, wherein about 51-100% of the glycosylated antibody, e.g., 80-100%, 90-100%, 95-100%, etc. of the antibody in the composition comprises a mature core carbohydrate structure which lacks fucose.
  • the antibody in the composition both comprises a mature core carbohydrate structure that lacks fucose and additionally comprises at least one amino acid modification in the Fc region.
  • a composition comprises a glycosylated immunoglobulin having an Fc region, wherein about 51-100% of the
  • glycosylated antibody, 80-100%, or 90-100%, of the antibody in the composition comprises a mature core carbohydrate structure which lacks sialic acid.
  • the antibody in the composition both comprises a mature core carbohydrate structure that lacks sialic acid and additionally comprises at least one amino acid modification in the Fc region.
  • a composition comprises a glycosylated immunoglobulin having an Fc region, wherein about 51 -100% of the glycosylated antibody, 80-100%, or 90-100%, of the antibody in the composition comprises a mature core carbohydrate structure which contains sialic acid.
  • the antibody in the composition both comprises a mature core carbohydrate structure that contains sialic acid and additionally comprises at least one amino acid modification in the Fc region.
  • the combination of engineered glycoform and amino acid modification provides optimal Fc receptor binding properties to the antibody.
  • Immunoglobulins disclosed herein may comprise one or more modifications that provide optimized properties. Said modifications may be amino acid modifications, or may be modifications that are made enzymatically or chemically. Such modification(s) likely provide some improvement in the immunoglobulin, for example an enhancement in its stability, solubility, function, or clinical use. Disclosed herein are a variety of improvements that may be made by coupling the immunoglobulins disclosed herein with additional modifications.
  • variable region of an immunoglobulin disclosed herein may be affinity matured, that is to say that amino acid modifications have been made in the VH and/or VL domains to enhance binding of the antibody to its target antigen. Such types of modifications may improve the association and/or the dissociation kinetics for binding to the target antigen. Other modifications include those that improve selectivity for target antigen vs. alternative targets. These include modifications that improve selectivity for antigen expressed on target vs. non-target cells. Immunoglobulins disclosed herein may comprise one or more modifications that provide reduced or enhanced internalization of an immunoglobulin disclosed herein.
  • modifications are made to improve biophysical properties of the immunoglobulins disclosed herein, including but not limited to stability, solubility, and oligomeric state.
  • Modifications can include, for example, substitutions that provide more favorable intramolecular interactions in the immunoglobulin such as to provide greater stability, or substitution of exposed nonpolar amino acids with polar amino acids for higher solubility.
  • Other modifications to the immunoglobulins disclosed herein include those that enable the specific formation or homodimeric or homomultimeric molecules. Such modifications include but are not limited to engineered disulfides, as well as chemical modifications or aggregation methods.
  • the immunoglobulins disclosed herein comprise
  • proteolytic degradation sites may include, for example, protease sites that reduce production yields, as well as protease sites that degrade the administered protein in vivo.
  • additional modifications are made to remove covalent degradation sites such as deamidation (i.e. deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues), oxidation, and proteolytic degradation sites.
  • Deamidation sites that are particular useful to remove are those that have enhance propensity for deamidation, including, but not limited to asparaginyl and gltuamyl residues followed by glycines (NG and QG motifs, respectively).
  • oxidation sites include methionine and cysteine residues.
  • Other covalent modifications that can either be introduced or removed, include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the "-amino groups of lysine, arginine, and histidine side chains, acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group. Additional modifications also may include but are not limited to posttranslational modifications such as N-linked or O-linked glycosylation and phosphorylation.
  • Modifications may include those that improve expression and/or purification yields from hosts or host cells commonly used for production of biologies. These include, but are not limited to various mammalian cell lines (e.g. CHO), yeast cell lines, bacterial cell lines, and plants. Additional modifications include modifications that remove or reduce the ability of heavy chains to form inter-chain disulfide linkages. Additional modifications include modifications that remove or reduce the ability of heavy chains to form intra-chain disulfide linkages.
  • the immunoglobulins disclosed herein may comprise modifications that include the use of unnatural amino acids incorporated using, including but not limited to methods described in Liu & Schultz, 2010, Annu Rev Biochem 79:413-444, herein expressly incorporated by reference. In some embodiments, these modifications enable manipulation of various functional, biophysical, immunological, or manufacturing properties discussed above. In additional embodiments, these modifications enable additional chemical modification for other purposes.
  • the immunoglobulin may be linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and
  • polypropylene glycol polypropylene glycol. Additional amino acid modifications may be made to enable specific or non-specific chemical or posttranslational modification of the immunoglobulins. Such modifications, include, but are not limited to PEGylation and glycosylation. Specific substitutions that can be utilized to enable PEGylation include, but are not limited to, introduction of novel cysteine residues or unnatural amino acids such that efficient and specific coupling chemistries can be used to attach a PEG or otherwise polymeric moiety. Introduction of specific glycosylation sites can be achieved by introducing novel N-X-T/S sequences into the immunoglobulins disclosed herein.
  • Modifications to reduce immunogenicity may include modifications that reduce binding of processed peptides derived from the parent sequence to MHC proteins. For example, amino acid modifications would be engineered such that there are no or a minimal number of immune epitopes that are predicted to bind, with high affinity, to any prevalent MHC alleles.
  • amino acid modifications would be engineered such that there are no or a minimal number of immune epitopes that are predicted to bind, with high affinity, to any prevalent MHC alleles.
  • Covalent modifications are included within the scope of immunoglobulins disclosed herein, and are generally, but not always, done post-translationally. For example, several types of covalent modifications can be introduced into the molecule by reacting specific amino acid residues with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues.
  • the covalent modification of the immunogloublins disclosed herein comprises the addition of one or more labels.
  • labeling group means any detectable label.
  • the labeling group is coupled to the immunoglobulin via spacer arms of various lengths to reduce potential steric hindrance.
  • Various methods for labeling proteins are known in the art and may be used in generating immunoglobulins disclosed herein.
  • the immunoglobulins disclosed herein are "fusion proteins", sometimes referred to herein as “conjugates".
  • the fusion partner or conjugate partner can be proteinaceous or non-proteinaceous; the latter generally being generated using functional groups on the immunoglobulin and on the conjugate partner.
  • Conjugate and fusion partners may be any molecule, including small molecule chemical compounds and polypeptides. For example, a variety of conjugates and methods are described in Trail et al., 1999, Curr. Opin. Immunol. 1 1 :584-588, incorporated entirely by reference. Possible conjugate partners include but are not limited to cytokines, cytotoxic agents, toxins, radioisotopes,
  • conjugate partners may be thought of more as payloads, that is to say that the goal of a conjugate is targeted delivery of the conjugate partner to a targeted cell, for example a cancer cell or immune cell, by the immunoglobulin.
  • the conjugation of a toxin to an immunoglobulin targets the delivery of said toxin to cells expressing the target antigen.
  • the concepts and definitions of fusion and conjugate are overlapping. The designation of a fusion or conjugate is not meant to constrain it to any particular embodiment disclosed herein. Rather, these terms are used loosely to convey the broad concept that any immunoglobulin disclosed herein may be linked genetically, chemically, or otherwise, to one or more polypeptides or molecules to provide some desirable property.
  • Suitable conjugates include, but are not limited to, labels as described below, drugs and cytotoxic agents including, but not limited to, cytotoxic drugs (e.g., chemotherapeutic agents) or toxins or active fragments of such toxins.
  • cytotoxic drugs e.g., chemotherapeutic agents
  • Suitable toxins and their corresponding fragments include diptheria A chain, exotoxin A chain, ricin A chain, abrin A chain, curcin, crotin, phenomycin, enomycin and the like.
  • Cytotoxic agents also include radiochemicals made by conjugating radioisotopes to immunoglobulin, or binding of a radionuclide to a chelating agent that has been covalently attached to the immunoglobulin. Additional embodiments utilize calicheamicin, auristatins, geldanamycin, maytansine, and
  • the immunoglobulins disclosed herein are fused or conjugated to a cytokine.
  • cytokine as used herein is meant a generic term for proteins released by one cell population that act on another cell as intercellular mediators.
  • cytokines may be fused to an immunoglobulin to provide an array of desirable properties.
  • lymphokines monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone;
  • parathyroid hormone parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin;
  • FSH follicle stimulating hormone
  • TSH thyroid stimulating hormone
  • LH luteinizing hormone
  • vascular endothelial growth factor integrin
  • thrombopoietin TPO
  • nerve growth factors such as NGF-beta; platelet-growth factor; transforming growth factors (TGFs) such as TGF-alpha and TGF-beta; insulin-like growth factor-l and -II; erythropoietin (EPO); osteoinductive factors; interferons such as interferon-alpha, beta, and -gamma; colony stimulating factors (CSFs) such as macrophage- CSF (M-CSF); granulocyte-macrophage-CSF (GM-CSF); and granulocyte-CSF (G-CSF); interleukins (ILs) such as IL-1 , IL-1 alpha, IL-2, IL
  • ILs interleukins
  • immunoglobulin-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g. avidin) which is conjugated to a cytotoxic agent (e.g. a radionucleotide).
  • a "ligand” e.g. avidin
  • cytotoxic agent e.g. a radionucleotide
  • the immunoglobulin is conjugated or operably linked to an enzyme in order to employ Antibody Dependent Enzyme Mediated Prodrug Therapy (ADEPT).
  • ADEPT may be used by conjugating or operably linking the immunoglobulin to a prodrug-activating enzyme that converts a prodrug (e.g. a peptidyl chemotherapeutic agent.
  • the disclosed methods are not meant to constrain embodiments to any particular application or theory of operation. Rather, the provided methods are meant to illustrate generally that one or more immunoglobulins may be produced and experimentally tested to obtain immunoglobulins.
  • General methods for antibody molecular biology, expression, purification, and screening are described in Antibody Engineering, edited by Kontermann & Dubel, Springer, Heidelberg, 2001 ; and Hayhurst & Georgiou, 2001 , Curr Opin Chem Biol 5:683-689; Maynard & Georgiou, 2000, Annu Rev Biomed Eng 2:339-76;
  • nucleic acids are created that encode the immunoglobulins, and that may then be cloned into host cells, expressed and assayed, if desired.
  • nucleic acids, and particularly DNA may be made that encode each protein sequence.
  • These practices are carried out using well-known procedures. For example, a variety of methods that may find use in generating immunoglobulins disclosed herein are described in Molecular Cloning - A Laboratory Manual, 3rd Ed. (Maniatis, Cold Spring Harbor Laboratory Press, New York, 2001 ), and Current Protocols in Molecular Biology (John Wiley & Sons), both incorporated entirely by reference. There are a variety of techniques that may be used to efficiently generate DNA encoding immunoglobulins disclosed herein.
  • Such methods include but are not limited to gene assembly methods, PCR-based method and methods which use variations of PCR, ligase chain reaction-based methods, pooled oligo methods such as those used in synthetic shuffling, error-prone amplification methods and methods which use oligos with random mutations, classical site- directed mutagenesis methods, cassette mutagenesis, and other amplification and gene synthesis methods.
  • gene assembly methods PCR-based method and methods which use variations of PCR
  • ligase chain reaction-based methods pooled oligo methods such as those used in synthetic shuffling
  • error-prone amplification methods and methods which use oligos with random mutations
  • classical site- directed mutagenesis methods cassette mutagenesis
  • cassette mutagenesis cassette mutagenesis
  • other amplification and gene synthesis methods include but are not limited to gene assembly methods, PCR-based method and methods which use variations of PCR, ligase chain reaction-based methods, pooled oligo methods such as those used in synthetic shuff
  • the immunoglobulin proteins disclosed herein may be produced by culturing a host cell transformed with nucleic acid, e.g., an expression vector, containing nucleic acid encoding the immunoglobulins, under the appropriate conditions to induce or cause expression of the protein.
  • the conditions appropriate for expression will vary with the choice of the expression vector and the host cell, and will be easily ascertained by one skilled in the art through routine experimentation.
  • a wide variety of appropriate host cells may be used, including but not limited to mammalian cells, bacteria, insect cells, yeast, and plant cells.
  • a variety of cell lines that may find use in generating immunoglobulins disclosed herein are described in the ATCC® cell line catalog, available from the American Type Culture Collection.
  • the immunoglobulins are expressed in mammalian expression systems, including systems in which the expression constructs are introduced into the mammalian cells using virus such as retrovirus or adenovirus.
  • virus such as retrovirus or adenovirus.
  • Any mammalian cells may be used, e.g., human, mouse, rat, hamster, and primate cells. Suitable cells also include known research cells, including but not limited to Jurkat T cells, NIH3T3, CHO, BHK, COS,
  • library proteins are expressed in bacterial cells.
  • Bacterial expression systems are well known in the art, and include Escherichia coli (E. coli), Bacillus subtilis, Streptococcus cremoris, and Streptococcus lividans.
  • immunoglobulins are produced in insect cells (e.g. Sf21/Sf9, Trichoplusia ni Bti-Tn5b1 -4) or yeast cells (e.g. S. cerevisiae, Pichia, etc).
  • immunoglobulins are expressed in vitro using cell free translation systems.
  • prokaryotic e.g. E. coli
  • eukaryotic e.g. wheat germ, rabbit reticulocytes
  • in vitro translation is required for some display technologies, for example ribosome display.
  • the immunoglobulins may be produced by chemical synthesis methods.
  • transgenic expression systems both animal (e.g. cow, sheep or goat milk, embryonated hen's eggs, whole insect larvae, etc.) and plant (e.g. corn, tobacco, duckweed, etc.)
  • the nucleic acids that encode the immunoglobulins disclosed herein may be incorporated into an expression vector in order to express the protein.
  • a variety of expression vectors may be utilized for protein expression.
  • Expression vectors may comprise self-replicating extra-chromosomal vectors or vectors which integrate into a host genome. Expression vectors are constructed to be compatible with the host cell type.
  • expression vectors which find use in generating immunoglobulins disclosed herein include but are not limited to those which enable protein expression in mammalian cells, bacteria, insect cells, yeast, and in in vitro systems.
  • a variety of expression vectors are available, commercially or otherwise, that may find use for expressing immunoglobulins disclosed herein.
  • Expression vectors typically comprise a protein operably linked with control or regulatory sequences, selectable markers, any fusion partners, and/or additional elements.
  • operably linked herein is meant that the nucleic acid is placed into a functional relationship with another nucleic acid sequence.
  • these expression vectors include transcriptional and translational regulatory nucleic acid operably linked to the nucleic acid encoding the immunoglobulin, and are typically appropriate to the host cell used to express the protein.
  • the transcriptional and translational regulatory sequences may include promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences.
  • expression vectors typically contain a selection gene or marker to allow the selection of transformed host cells containing the expression vector. Selection genes are well known in the art and will vary with the host cell used.
  • Immunoglobulins may be operably linked to a fusion partner to enable targeting of the expressed protein, purification, screening, display, and the like. Fusion partners may be linked to the immunoglobulin sequence via a linker sequences.
  • the linker sequence will generally comprise a small number of amino acids, typically less than ten, although longer linkers may also be used. Typically, linker sequences are selected to be flexible and resistant to degradation. As will be appreciated by those skilled in the art, any of a wide variety of sequences may be used as linkers.
  • a common linker sequence comprises the amino acid sequence GGGGS.
  • a fusion partner may be a targeting or signal sequence that directs immunoglobulin and any associated fusion partners to a desired cellular location or to the extracellular media.
  • fusion partner may also be a sequence that encodes a peptide or protein that enables purification and/or screening.
  • fusion partners include but are not limited to polyhistidine tags (His- tags) (for example H6 and H 10 or other tags for use with Immobilized Metal Affinity
  • IMAC Chromatography
  • GST fusions e.g. Ni+2 affinity columns
  • MBP fusions e.g. GST fusions
  • Strep-tag e.g. GST fusions
  • BSP biotinylation target sequence of the bacterial enzyme BirA e.g. c-myc tags, flag-tags, and the like
  • epitope tags which are targeted by antibodies (for example c-myc tags, flag-tags, and the like).
  • such tags may be useful for purification, for screening, or both.
  • an immunoglobulin may be purified using a His-tag by immobilizing it to a Ni+2 affinity column, and then after purification the same His-tag may be used to immobilize the antibody to a Ni+2 coated plate to perform an ELISA or other binding assay (as described below).
  • a fusion partner may enable the use of a selection method to screen immunoglobulins (see below). Fusion partners that enable a variety of selection methods are well-known in the art. [191] For example, by fusing the members of an immunoglobulin library to the gene III protein, phage display can be employed. Fusion partners may enable immunoglobulins to be labeled. Alternatively, a fusion partner may bind to a specific sequence on the expression vector, enabling the fusion partner and associated immunoglobulin to be linked covalently or noncovalently with the nucleic acid that encodes them. The methods of introducing exogenous nucleic acid into host cells are well known in the art, and will vary with the host cell used. Techniques include but are not limited to dextran-mediated transfection, calcium phosphate precipitation, calcium chloride treatment, polybrene mediated transfection, protoplast fusion, electroporation, viral or phage infection, encapsulation of the
  • transfection may be either transient or stable.
  • immunoglobulins are purified or isolated after expression.
  • Proteins may be isolated or purified in a variety of ways known to those skilled in the art. Purification may be particularly useful in the invention for separating heterodimeric heavy chain species from homodimeric heavy chain species, as described herein. Standard purification methods include chromatographic techniques, including ion exchange, hydrophobic interaction, affinity, sizing or gel filtration, and reversed-phase, carried out at atmospheric pressure or at high pressure using systems such as FPLC and HPLC.
  • Purification methods also include electrophoretic, isoelectric focusing, immunological, precipitation, dialysis, and chromatofocusing techniques. Ultrafiltration and diafiltration techniques, in conjunction with protein concentration, are also useful.
  • a variety of natural proteins bind Fc and antibodies, and these proteins can find use for purification of immunoglobulins disclosed herein.
  • the bacterial proteins A and G bind to the Fc region.
  • the bacterial protein L binds to the Fab region of some antibodies, as of course does the antibody's target antigen. Purification can often be enabled by a particular fusion partner.
  • immunoglobulins may be purified using glutathione resin if a GST fusion is employed, Ni+2 affinity chromatography if a His-tag is employed, or immobilized anti-flag antibody if a flag-tag is used.
  • glutathione resin if a GST fusion is employed
  • Ni+2 affinity chromatography if a His-tag is employed
  • immobilized anti-flag antibody if a flag-tag is used.
  • Immunoglobulins may be screened using a variety of in vitro methods, including but not limited to those that use binding assays, cell-based assays, and selection technologies. Automation and high-throughput screening technologies may be utilized in the screening procedures. Screening may employ the use of a fusion partner or label. The use of fusion partners has been discussed above.
  • label herein is meant that the immunoglobulins disclosed herein have one or more elements, isotopes, or chemical compounds attached to enable the detection in a screen.
  • labels fall into three classes: a) immune labels, which may be an epitope incorporated as a fusion partner that is recognized by an antibody, b) isotopic labels, which may be radioactive or heavy isotopes, and c) small molecule labels, which may include fluorescent and colorimetric dyes, or molecules such as biotin that enable other labeling methods. Labels may be incorporated into the compound at any position and may be incorporated in vitro or in vivo during protein expression.
  • the functional and/or biophysical properties of immunoglobulins are screened in an in vitro assay.
  • In vitro assays may allow a broad dynamic range for screening properties of interest. Particularly relevant for the present invention, the
  • immunoglobulins may be tested for their affinity for one or more antigens.
  • Properties that may be screened include but are not limited to stability, solubility, and affinity for Fc ligands, for example FcyRs. Multiple properties may be screened simultaneously or individually.
  • Proteins may be purified or unpurified, depending on the requirements of the assay.
  • the screen is a qualitative or quantitative binding assay for binding of immunoglobulins to a protein or nonprotein molecule that is known or thought to bind the immunoglobulin.
  • the screen is a binding assay for measuring binding to the target antigen.
  • the screen is an assay for binding of immunoglobulins to an Fc ligand, including but are not limited to the family of FcyRs, the neonatal receptor FcRn, the complement protein C1 q, and the bacterial proteins A and G.
  • Said Fc ligands may be from any organism.
  • Fc ligands are from humans, mice, rats, rabbits, and/or monkeys. Binding assays can be carried out using a variety of methods known in the art, including but not limited to FRET (Fluorescence
  • immunoglobulin may employ a variety of detection methods including but not limited to chromogenic, fluorescent, luminescent, or isotopic labels.
  • the biophysical properties of immunoglobulins may be tested using a variety of methods known in the art. Protein stability may be determined by measuring the thermodynamic equilibrium between folded and unfolded states. For example, immunoglobulins disclosed herein may be unfolded using chemical denaturant, heat, or pH, and this transition may be monitored using methods including but not limited to circular dichroism spectroscopy, fluorescence spectroscopy, absorbance spectroscopy, NMR spectroscopy, calorimetry, and proteolysis.
  • the kinetic parameters of the folding and unfolding transitions may also be monitored using these and other techniques.
  • the solubility and overall structural integrity of an immunoglobulin may be quantitatively or qualitatively determined using a wide range of methods that are known in the art.
  • Methods which may find use for characterizing the biophysical properties of immunoglobulins disclosed herein include gel electrophoresis, isoelectric focusing, capillary electrophoresis, chromatography such as size exclusion chromatography, ion-exchange chromatography, and reversed-phase high performance liquid chromatography, peptide mapping, oligosaccharide mapping, mass spectrometry, ultraviolet absorbance spectroscopy, fluorescence spectroscopy, circular dichroism spectroscopy, isothermal titration calorimetry, differential scanning calorimetry, analytical ultra-centrifugation, dynamic light scattering, proteolysis, and cross-linking, turbidity measurement, filter retardation assays, immunological assays, fluorescent dye binding assays, protein-staining assays, microscopy, and detection of aggregates via ELISA or other binding assay.
  • Structural analysis employing X-ray crystallographic techniques and NMR spectroscopy may also find use.
  • stability and/or solubility may be measured by determining the amount of protein solution after some defined period of time.
  • the protein may or may not be exposed to some extreme condition, for example elevated temperature, low pH, or the presence of denaturant.
  • the aforementioned functional and binding assays also provide ways to perform such a measurement. For example, a solution comprising an immunoglobulin could be assayed for its ability to bind target antigen, then exposed to elevated temperature for one or more defined periods of time, then assayed for antigen binding again. Because unfolded and aggregated protein is not expected to be capable of binding antigen, the amount of activity remaining provides a measure of the immunoglobulin's stability and solubility.
  • immunoglobulins may be tested using one or more cell-based or in vitro assays.
  • immunoglobulins purified or unpurified, are typically added exogenously such that cells are exposed to immunoglobulins described herein.
  • These assays are typically, but not always, based on the biology of the ability of the immunoglobulin to bind to the target antigen and mediate some biochemical event, for example effector functions like cellular lysis, phagocytosis, ligand/receptor binding inhibition, inhibition of growth and/or proliferation, inhibition of calcium release and/or signaling, apoptosis and the like.
  • Such assays often involve monitoring the response of cells to immunoglobulin, for example cell survival, cell death, cellular phagocytosis, cell lysis, change in cellular morphology, or transcriptional activation such as cellular expression of a natural gene or reporter gene.
  • immunoglobulin for example cell survival, cell death, cellular phagocytosis, cell lysis, change in cellular morphology, or transcriptional activation such as cellular expression of a natural gene or reporter gene.
  • such assays may measure the ability of
  • immunoglobulins to elicit cell killing for example ADCC, ADCP, and CDC.
  • Assays that measure cellular killing that is mediated by co-engagement of antigens are particularly relevant for the invention.
  • additional cells or components, that is in addition to the target cells may need to be added, for example serum complement, or effector cells such as peripheral blood monocytes (PBMCs), NK cells, macrophages, T cells, and the like.
  • PBMCs peripheral blood monocytes
  • NK cells NK cells
  • macrophages T cells
  • additional cells may be from any organism, e.g., humans, mice, rat, rabbit, and monkey.
  • Crosslinked or monomeric antibodies may cause apoptosis of certain cell lines expressing the antibody's target antigen, or they may mediate attack on target cells by immune cells which have been added to the assay.
  • Methods for monitoring cell death or viability include the use of dyes, fluorophores, immunochemical, cytochemical, and radioactive reagents.
  • caspase assays or annexin- flourconjugates may enable apoptosis to be measured, and uptake or release of radioactive substrates (e.g. Chromium-51 release assays) or the metabolic reduction of fluorescent dyes such as alamar blue may enable cell growth, proliferation or activation to be monitored.
  • the DELFIA EuTDA-based cytotoxicity assay (Perkin Elmer, MA) is used.
  • dead or damaged target cells may be monitored by measuring the release of one or more natural intracellular proteins, for example lactate dehydrogenase.
  • Transcriptional activation may also serve as a method for assaying function in cell-based assays.
  • response may be monitored by assaying for natural genes or proteins which may be upregulated or down-regulated, for example the release of certain interleukins may be measured, or alternatively readout may be via a luciferase or GFP-reporter construct.
  • Cell-based assays may also involve the measure of morphological changes of cells as a response to the presence of an immunoglobulin.
  • Cell types for such assays may be prokaryotic or eukaryotic, and a variety of cell lines that are known in the art may be employed.
  • cell-based screens are performed using cells that have been transformed or transfected with nucleic acids encoding the immunoglobulins.
  • drugs are often tested in animals, including but not limited to mice, rats, rabbits, dogs, cats, pigs, and monkeys, in order to measure a drug's efficacy for treatment against a disease or disease model, or to measure a drug's pharmacokinetics, toxicity, and other properties. Said animals may be referred to as disease models.
  • Therapeutics are often tested in mice, including but not limited to nude mice, Rag-deficient mice, SCID mice, xenograft mice, and transgenic mice (including knockins and knockouts).
  • an immunoglobulin of the present invention that is intended as an anti-cancer therapeutic may be tested in a mouse cancer model, for example a xenograft mouse.
  • a tumor or tumor cell line is grafted onto or injected into a mouse, and subsequently the mouse is treated with the therapeutic to determine the ability of the drug to reduce or inhibit cancer growth and metastasis.
  • Therapeutic immunoglobulins herein can be tested in mouse strains NZB, NOD, BXSB, MRL/lpr, K/BxN and transgenics (including knockins and knockouts). Such mice can develop various autoimmune conditions that resemble human organ specific, systemic autoimmune or inflammatory disease pathologies such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA).
  • SLE systemic lupus erythematosus
  • RA rheumatoid arthritis
  • an immunoglobulin disclosed herein intended for autoimmune diseases may be tested in such mouse models by treating the mice to determine the ability of the immunoglobulin to reduce or inhibit the development of the disease pathology.
  • an alternative approach is to use a murine SCID model in which immune deficient mice are engrafted with human PBLs or PBMCs (huPBL-SCID, huPBMC-SCID) providing a semi-functional human immune system with human effector cells and Fc receptors.
  • Other organisms e.g., mammals, may also be used for testing.
  • monkeys can be suitable therapeutic models, and thus may be used to test the efficacy, toxicity, pharmacokinetics, or other property of the immunoglobulins disclosed herein.
  • immunoglobulins disclosed herein may be tested in humans to determine their therapeutic efficacy, toxicity, pharmacokinetics, and/or other clinical properties.
  • immunoglobulins disclosed herein may be assessed for efficacy in clinically relevant animal models of various human diseases. In many cases, relevant models include various transgenic animals for specific antigens and receptors.
  • the testing of immunoglobulins may include study of efficacy in primates (e.g. cynomolgus monkey model) to facilitate the evaluation of depletion of specific target cells harboring the target antigen.
  • primates e.g. cynomolgus monkey model
  • Additional primate models include but are not limited to use of the rhesus monkey to assess immunoglobulins in therapeutic studies of autoimmune, transplantation and cancer.
  • Toxicity studies are performed to determine drug related-effects that cannot be evaluated in standard pharmacology profiles, or occur only after repeated administration of the agent. Most toxicity tests are performed in two species - a rodent and a non-rodent - to ensure that any unexpected adverse effects are not overlooked before new therapeutic entities are introduced into man. In general, these models may measure a variety of toxicities including genotoxicity, chronic toxicity, immunogenicity,
  • reproductive/developmental toxicity and carcinogenicity include reproductive/developmental toxicity and carcinogenicity. Included within the aforementioned parameters are standard measurement of food consumption, bodyweight, antibody formation, clinical chemistry, and macro- and microscopic examination of standard organs/tissues (e.g. cardiotoxicity). Additional parameters of measurement are injection site trauma and the measurement of neutralizing antibodies, if any. Traditionally, monoclonal antibody therapeutics, naked or conjugated, is evaluated for cross-reactivity with normal tissues, immunogenicity/antibody production, conjugate or linker toxicity and "bystander" toxicity of radiolabeled species. Nonetheless, such studies may have to be individualized to address specific concerns and following the guidance set by ICH S6 (Safety studies for biotechnological products, also noted above). As such, the general principles are that the products are sufficiently well characterized, impurities/contaminants have been removed, that the test material is comparable throughout development, and that GLP compliance is maintained.
  • PK pharmacokinetics
  • the pharmacokinetics (PK) of the immunoglobulins disclosed herein may be studied in a variety of animal systems, with the most relevant being non-human primates such as the cynomolgus and rhesus monkeys.
  • Single or repeated i.v./s.c. administrations over a dose range of 6000-fold (0.05-300 mg/kg) can be evaluated for half-life (days to weeks) using plasma concentration and clearance. Volume of distribution at a steady state and level of systemic absorbance can also be measured.
  • Examples of such parameters of measurement generally include maximum observed plasma concentration (Cmax), the time to reach Cmax (Tmax), the area under the plasma concentration-time curve from time 0 to infinity [AUC(0- inf] and apparent elimination half-life (T1 /2). Additional measured parameters could include compartmental analysis of concentration-time data obtained following i.v. administration and bioavailability.
  • Pharmacodynamic studies may include, but are not limited to, targeting specific cells or blocking signaling mechanisms, measuring inhibition of antigen-specific antibodies etc.
  • the immunoglobulins disclosed herein may target particular effector cell populations and thereby be direct drugs to induce certain activities to improve potency or to increase penetration into a particularly favorable physiological compartment. Such pharmacodynamic effects may be demonstrated in animal models or in humans.
  • the immunoglobulins disclosed herein may find use in a wide range of products.
  • an immunoglobulin disclosed herein is a therapeutic, a diagnostic, or a research reagent.
  • the immunoglobulins may find use in a composition that is monoclonal or polyclonal.
  • the immunoglobulins disclosed herein may be used for therapeutic purposes.
  • the immunoglobulins disclosed herein may be used for any therapeutic purpose that antibodies, Fc fusions, and the like may be used for.
  • the immunoglobulins may be administered to a patient to treat disorders including but not limited to cancer, infectious diseases, autoimmune and inflammatory diseases.
  • a "patient” for the purposes disclosed herein includes both humans and other animals, e.g., other mammals.
  • the immunoglobulins disclosed herein have both human therapy and veterinary applications.
  • treatment or “treating” as disclosed herein is meant to include therapeutic treatment, as well as prophylactic, or suppressive measures for a disease or disorder.
  • successful administration of an immunoglobulin prior to onset of the disease results in treatment of the disease.
  • successful administration of an optimized immunoglobulin after clinical manifestation of the disease to combat the symptoms of the disease comprises treatment of the disease.
  • Treatment and “treating” also encompasses administration of an optimized immunoglobulin after the appearance of the disease in order to eradicate the disease.
  • Those "in need of treatment” include mammals already having the disease or disorder, as well as those prone to having the disease or disorder, including those in which the disease or disorder is to be prevented.
  • an immunoglobulin disclosed herein is administered to a patient having a disease involving inappropriate expression of a protein or other molecule.
  • this is meant to include diseases and disorders characterized by aberrant proteins, due for example to alterations in the amount of a protein present, protein localization, posttranslational modification, conformational state, the presence of a mutant or pathogen protein, etc.
  • the disease or disorder may be characterized by alterations molecules including but not limited to polysaccharides and gangliosides.
  • An overabundance may be due to any cause, including but not limited to overexpression at the molecular level, prolonged or accumulated appearance at the site of action, or increased activity of a protein relative to normal.
  • diseases and disorders characterized by a reduction of a protein.
  • This reduction may be due to any cause, including but not limited to reduced expression at the molecular level, shortened or reduced appearance at the site of action, mutant forms of a protein, or decreased activity of a protein relative to normal.
  • Such an overabundance or reduction of a protein can be measured relative to normal expression, appearance, or activity of a protein, and said measurement may play an important role in the development and/or clinical testing of the immunoglobulins disclosed herein.
  • the immunoglobulins herein may be used to treat cancer.
  • cancer By “cancer” and
  • cancer herein refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • examples of cancer include but are not limited to carcinoma, lymphoma, blastoma, sarcoma (including liposarcoma),
  • neuroendocrine tumors mesothelioma, schwanoma, meningioma, adenocarcinoma, melanoma, and leukemia or lymphoid malignancies.
  • cancers include hematologic malignancies, such as Hodgkin's lymphoma; non-Hodgkin's lymphomas (Burkitt's lymphoma, small lymphocytic lymphoma/chronic lymphocytic leukemia, mycosis fungoides, mantle cell lymphoma, follicular lymphoma, diffuse large B-cell lymphoma, marginal zone lymphoma, hairy cell leukemia and lymphoplasmacytic leukemia), tumors of lymphocyte precursor cells, including B-cell acute lymphoblastic leukemia/lymphoma, and T-cell acute lymphoblastic
  • leukemia/lymphoma thymoma
  • tumors of the mature T and NK cells including peripheral T- cell leukemias, adult T-cell leukemia/T-cell lymphomas and large granular lymphocytic leukemia, Langerhans cell histocytosis, myeloid neoplasias such as acute myelogenous leukemias, including AML with maturation, AML without differentiation, acute promyelocytic leukemia, acute myelomonocytic leukemia, and acute monocytic leukemias, myelodysplastic syndromes, and chronic myeloproliferative disorders, including chronic myelogenous leukemia; tumors of the central nervous system such as glioma, glioblastoma,
  • neuroblastoma astrocytoma, medulloblastoma, ependymoma, and retinoblastoma
  • solid tumors of the head and neck eg. nasopharyngeal cancer, salivary gland carcinoma, and esophagael cancer
  • lung eg. small-cell lung cancer, non-small cell lung cancer
  • adenocarcinoma of the lung and squamous carcinoma of the lung digestive system
  • digestive system eg. gastric or stomach cancer including gastrointestinal cancer, cancer of the bile duct or biliary tract, colon cancer, rectal cancer, colorectal cancer, and anal carcinoma
  • reproductive system eg. testicular, penile, or prostate cancer, uterine, vaginal, vulval, cervical, ovarian, and endometrial cancer
  • skin eg. melanoma, basal cell carcinoma, squamous cell cancer, actinic keratosis
  • liver eg. liver cancer, hepatic carcinoma, hepatocellular cancer, and hepatoma
  • bone eg.
  • osteoclastoma and osteolytic bone cancers
  • additional tissues and organs eg. pancreatic cancer, bladder cancer, kidney or renal cancer, thyroid cancer, breast cancer, cancer of the peritoneum, and Kaposi's sarcoma
  • tumors of the vascular system eg. angiosarcoma and hemagiopericytoma.
  • the immunoglobulins herein may be used to treat autoimmune diseases.
  • autoimmune diseases herein include allogenic islet graft rejection, alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, autoimmune Addison's disease, antineutrophil cytoplasmic autoantibodies (ANCA), autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune myocarditis, autoimmune neutropenia, autoimmune oophoritis and orchitis, autoimmune
  • thrombocytopenia thrombocytopenia, autoimmune urticaria, Behcet's disease, bullous pemphigoid,
  • cardiomyopathy Castleman's syndrome, celiac spruce-dermatitis, chronic fatigue immune disfunction syndrome, chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, cicatrical pemphigoid, CREST syndrome, cold agglutinin disease, Crohn's disease, dermatomyositis, discoid lupus, essential mixed cryoglobulinemia, factor VIII deficiency, fibromyalgia-fibromyositis, glomerulonephritis, Grave's disease, Guillain-Barre, Goodpasture's syndrome, graft-versus-host disease (GVHD), Hashimoto's thyroiditis, hemophilia A, idiopathic pulmonary fibrosis, idiopathic thrombocytopenia purpura (ITP), IgA neuropathy, IgM polyneuropathies, immune mediated thrombocytopenia, juvenile arthritis, Kawasaki's disease,
  • agammaglobinulinemia primary biliary cirrhosis, psoriasis, psoriatic arthritis, Reynauld's phenomenon, Reiter's syndrome, rheumatoid arthritis, sarcoidosis, scleroderma, Sjorgen's syndrome, solid organ transplant rejection, stiff-man syndrome, systemic lupus
  • erythematosus erythematosus, takayasu arteritis, temporal arteristis / giant cell arteritis, thrombotic thrombocytopenia purpura, ulcerative colitis, uveitis, vasculitides such as dermatitis herpetiformis vasculitis, vitiligo, and Wegner's granulomatosis.
  • the immunoglobulins herein may be used to treat inflammtory disorders.
  • inflammatory disorders herein include acute respiratory distress syndrome (ARDS), acute septic arthritis, adjuvant arthritis, juvenile idiopathic arthritis, allergic encephalomyelitis, allergic rhinitis, allergic vasculitis, allergy, asthma, atherosclerosis, chronic inflammation due to chronic bacterial or viral infectionis, chronic obstructive pulmonary disease (COPD), coronary artery disease, encephalitis, inflammatory bowel disease, inflammatory osteolysis, inflammation associated with acute and delayed hypersensitivity reactions, inflammation associated with tumors, peripheral nerve injury or demyelinating diseases, inflammation associated with tissue trauma such as burns and ischemia, inflammation due to meningitis, multiple organ injury syndrome, pulmonary fibrosis, sepsis and septic shock, Stevens- Johnson syndrome, undifferentiated arthropy, and undifferentiated spondyloarthropathy.
  • ARDS acute respiratory distress syndrome
  • COPD chronic obstructive pulmonary disease
  • COPD chronic obstructive pulmonary disease
  • coronary artery disease
  • Some autoimmune and inflammatory diseases that may be targeted by the immunoglobulins disclosed herein include Systemic Lupus Erythematosus, Rheumatoid arthritis, Sjogren's syndrome, Multiple sclerosis, Idiopathic thrombocytopenic purpura (ITP), Graves disease, Inflammatory bowel disease, Psoriasis, Type I diabetes, and Asthma.
  • infectious diseases include diseases caused by pathogens such as viruses, bacteria, fungi, protozoa, and parasites. Infectious diseases may be caused by viruses including adenovirus, cytomegalovirus, dengue, Epstein-Barr, hanta, hepatitis A, hepatitis B, hepatitis C, herpes simplex type I, herpes simplex type II, human immunodeficiency virus, (HIV), human papilloma virus (HPV), influenza, measles, mumps, papova virus, polio, respiratory syncytial virus, rinderpest, rhinovirus, rotavirus, rubella, SARS virus, smallpox, viral meningitis, and the like.
  • infectious diseases include diseases caused by pathogens such as viruses, bacteria, fungi, protozoa, and parasites. Infectious diseases may be caused by viruses including adenovirus, cytomegalovirus, dengue, Epstein-Barr,
  • Infections diseases may also be caused by bacteria including Bacillus antracis, Borrelia burgdorferi, Campylobacter jejuni, Chlamydia trachomatis, Clostridium botulinum, Clostridium tetani, Diptheria, E. coli, Legionella, Helicobacter pylori, Mycobacterium rickettsia, Mycoplasma nesisseria, Pertussis, Pseudomonas aeruginosa, S. pneumonia, Streptococcus, Staphylococcus, Vibria cholerae, Yersinia pestis, and the like. Infectious diseases may also be caused by fungi such as Aspergillus fumigatus, Blastomyces dermatitidis, Candida albicans, Coccidioides immitis, Cryptococcus neoformans,
  • Infectious diseases may also be caused by protozoa and parasites such as chlamydia, kokzidioa, leishmania, malaria, rickettsia, trypanosoma, and the like.
  • immunoglobulins disclosed herein may be used to prevent or treat additional conditions including but not limited to heart conditions such as congestive heart failure (CHF), myocarditis and other conditions of the myocardium; skin conditions such as rosecea, acne, and eczema; bone and tooth conditions such as bone loss, osteoporosis, Paget's disease, Langerhans' cell histiocytosis, periodontal disease, disuse osteopenia, osteomalacia, monostotic fibrous dysplasia, polyostotic fibrous dysplasia, bone metastasis, bone pain management, humoral malignant hypercalcemia, periodontal reconstruction, spinal cord injury, and bone fractures; metabolic conditions such as Gaucher's disease; endocrine conditions such as Cushing's syndrome; and neurological and neurodegenerative conditions such as Alzheimer's disease.
  • CHF congestive heart failure
  • myocarditis and other conditions of the myocardium skin conditions such as rosecea, acne, and eczema
  • compositions are contemplated wherein an immunoglobulin disclosed herein and one or more therapeutically active agents are formulated.
  • Formulations of the immunoglobulins disclosed herein are prepared for storage by mixing said
  • immunoglobulin having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed., 1980, incorporated entirely by reference), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, acetate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride;
  • hexamethonium chloride benzalkonium chloride, benzethonium chloride; phenol, butyl orbenzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; sweeteners and other flavoring agents; fillers such as microcrystalline cellulose
  • the pharmaceutical composition that comprises the immunoglobulin disclosed herein may be in a water-soluble form, such as being present as pharmaceutically acceptable salts, which is meant to include both acid and base addition salts.
  • pharmaceutically acceptable acid addition salt refers to those salts that retain the biological effectiveness of the free bases and that are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • “Pharmaceutically acceptable base addition salts” include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Some embodiments include at least one of the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
  • the formulations to be used for in vivo administration may be sterile. This is readily accomplished by filtration through sterile filtration membranes or other methods.
  • the immunoglobulins disclosed herein may also be formulated as immunoliposomes.
  • a liposome is a small vesicle comprising various types of lipids, phospholipids and/or surfactant that is useful for delivery of a therapeutic agent to a mammal.
  • Liposomes containing the immunoglobulin are prepared by methods known in the art. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising
  • Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • the immunoglobulin and other therapeutically active agents may also be entrapped in microcapsules prepared by methods including but not limited to coacervation techniques, interfacial polymerization (for example using hydroxymethylcellulose or gelatin- microcapsules, or poly-(methylmethacylate) microcapsules), colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules), and macroemulsions.
  • coacervation techniques for example using hydroxymethylcellulose or gelatin- microcapsules, or poly-(methylmethacylate) microcapsules
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymer, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (for example poly(2- hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and gamma ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the Lupron Depot® (which are injectable
  • microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), poly- D-(-)-3-hydroxybutyric acid, and ProLease® (commercially available from Alkermes), which is a microsphere-based delivery system composed of the desired bioactive molecule incorporated into a matrix of poly-DL-lactide-co-glycolide (PLG).
  • PLG poly-DL-lactide-co-glycolide
  • Administration of the pharmaceutical composition comprising an immunoglobulin disclosed herein may be done in a variety of ways, including, but not limited to orally, subcutaneously, intravenously, intranasally, intraotically, transdermally, topically (e.g., gels, salves, lotions, creams, etc.),
  • the immunoglobulin may be directly applied as a solution or spray.
  • the pharmaceutical composition may be formulated accordingly depending upon the manner of introduction.
  • Subcutaneous administration may be used in circumstances where the patient may self-administer the pharmaceutical composition.
  • Many protein therapeutics are not sufficiently potent to allow for formulation of a therapeutically effective dose in the maximum acceptable volume for subcutaneous administration. This problem may be addressed in part by the use of protein formulations comprising arginine-HCI, histidine, and polysorbate.
  • Immunoglobulins disclosed herein may be more amenable to subcutaneous administration due to, for example, increased potency, improved serum half-life, or enhanced solubility.
  • protein therapeutics are often delivered by IV infusion or bolus.
  • the immunoglobulins disclosed herein may also be delivered using such methods.
  • administration may be by intravenous infusion with 0.9% sodium chloride as an infusion vehicle.
  • Pulmonary delivery may be accomplished using an inhaler or nebulizer and a formulation comprising an aerosolizing agent.
  • an inhaler or nebulizer for example, AERx® inhalable technology commercially available from Aradigm, or InhanceTM pulmonary delivery system commercially available from Nektar Therapeutics may be used.
  • immunoglobulins disclosed herein may be amenable to oral delivery.
  • any of a number of delivery systems are known in the art and may be used to administer the immunoglobulins disclosed herein. Examples include, but are not limited to, encapsulation in liposomes, microparticles, microspheres (e.g., PLA PGA microspheres), and the like.
  • an implant of a porous, non-porous, or gelatinous material, including membranes or fibers may be used.
  • Sustained release systems may comprise a polymeric material or matrix such as polyesters, hydrogels,
  • a nucleic acid encoding an immunoglobulin disclosed herein for example by retroviral infection, direct injection, or coating with lipids, cell surface receptors, or other transfection agents.
  • controlled release systems may be used to release the immunoglobulin at or close to the desired location of action.
  • the dosing amounts and frequencies of administration are, in one embodiment, selected to be therapeutically or prophylactically effective.
  • adjustments for protein degradation, systemic versus localized delivery, and rate of new protease synthesis, as well as the age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition may be necessary, and will be ascertainable with routine experimentation by those skilled in the art.
  • the concentration of the therapeutically active immunoglobulin in the formulation may vary from about 0.1 to 100 weight %. In one embodiment, the concentration of the immunoglobulin is in the range of 0.003 to 1 .0 molar. In order to treat a patient, a
  • therapeutically effective dose of the immunoglobulin disclosed herein may be administered.
  • therapeutically effective dose herein is meant a dose that produces the effects for which it is administered. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques. Dosages may range from 0.0001 to 100 mg/kg of body weight or greater, for example 0.1 , 1 , 10, or 50 mg/kg of body weight. In one embodiment, dosages range from 1 to 10mg/kg.
  • only a single dose of the immunoglobulin is used. In other embodiments, multiple doses of the immunoglobulin are administered.
  • the elapsed time between administrations may be less than 1 hour, about 1 hour, about 1-2 hours, about 2-3 hours, about 3-4 hours, about 6 hours, about 12 hours, about 24 hours, about 48 hours, about 2-4 days, about 4-6 days, about 1 week, about 2 weeks, or more than 2 weeks.
  • the immunoglobulins disclosed herein are administered in metronomic dosing regimes, either by continuous infusion or frequent administration without extended rest periods. Such metronomic administration may involve dosing at constant intervals without rest periods. Typically such regimens encompass chronic low-dose or continuous infusion for an extended period of time, for example 1 -2 days, 1-2 weeks, 1-2 months, or up to 6 months or more. The use of lower doses may minimize side effects and the need for rest periods.
  • the immunoglobulins disclosed herein and one or more other prophylactic or therapeutic agents are cyclically administered to the patient.
  • Cycling therapy involves administration of a first agent at one time, a second agent at a second time, optionally additional agents at additional times, optionally a rest period, and then repeating this sequence of administration one or more times.
  • the number of cycles is typically from 2 - 10. Cycling therapy may reduce the development of resistance to one or more agents, may minimize side effects, or may improve treatment efficacy.
  • the immunoglobulins disclosed herein may be administered concomitantly with one or more other therapeutic regimens or agents.
  • the additional therapeutic regimes or agents may be used to improve the efficacy or safety of the immunoglobulin.
  • the additional therapeutic regimes or agents may be used to treat the same disease or a comorbidity rather than to alter the action of the immunoglobulin.
  • an immunoglobulin disclosed herein may be administered to the patient along with chemotherapy, radiation therapy, or both chemotherapy and radiation therapy.
  • immunoglobulins disclosed herein and the other agent or agents are administered in a sequence and within a time interval such that they may act together to provide a benefit that is increased versus treatment with only either the immunoglobulin disclosed herein or the other agent or agents.
  • immunoglobulins disclosed herein and the other agent or agents act additively, and sometimes
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the skilled medical practitioner can determine empirically, or by considering the pharmacokinetics and modes of action of the agents, the appropriate dose or doses of each therapeutic agent, as well as the appropriate timings and methods of administration.
  • the immunoglobulin disclosed herein may be administered in combination with one or more other prophylactic or therapeutic agents, including but not limited to cytotoxic agents, chemotherapeutic agents, antibiotics, antifungal agents, antiviral agents, cytokines, growth inhibitory agents, anti-hormonal agents, kinase inhibitors, anti-angiogenic agents, cardioprotectants, immunostimulatory agents, immunosuppressive agents, agents that promote proliferation of hematological cells, angiogenesis inhibitors, protein tyrosine kinase (PTK) inhibitors, other antibodies, Fc fusions, or immunoglobulins, or other therapeutic agents.
  • the therapies of the invention may be combined with other immunotherapies.
  • the therapies of the invention may be combined with antagonists of chemokines or cytokines, including but not limited to antibodies and Fc fusions.
  • immunoglobulins disclosed herein may be combined with other therapeutic regimens.
  • immunoglobulin disclosed herein may also receive radiation therapy.
  • Radiation therapy can be administered according to protocols commonly employed in the art and known to the skilled artisan. Such therapy includes but is not limited to cesium, iridium, iodine, or cobalt radiation.
  • the radiation therapy may be whole body irradiation, or may be directed locally to a specific site or tissue in or on the body, such as the lung, bladder, or prostate.
  • the radiation therapy may be administered as a single dose or as multiple, sequential doses. The skilled medical practitioner can determine empirically the appropriate dose or doses of radiation therapy useful herein.
  • an immunoglobulin disclosed herein and one or more other anti-cancer therapies are employed to treat cancer cells ex vivo.
  • ex vivo treatment may be useful in bone marrow transplantation and particularly, autologous bone marrow transplantation.
  • treatment of cells or tissue(s) containing cancer cells with immunoglobulin and one or more other anti-cancer therapies, such as described above can be employed to deplete or substantially deplete the cancer cells prior to transplantation in a recipient patient.
  • the immunoglobulins disclosed herein may employ in combination with still other therapeutic techniques such as surgery.
  • scFv-Fc and empty-Fc constructs with native lgG1 Fc regions were cotransfected into HEK293E cells (Durocher et al., 2002, Nucleic Acids Research 30: E9) for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography.
  • the protein species generated were visualized electrophoretically using an Agilent 2100 Bioanalyzer microfluidics-based platform (Agilent Technologies) .
  • Agilent chips for SDS-PAGE were run on the BioAnalyzer using kit# 5067-1517, lot# NA06BP01 containing gels, chips, and buffers, carried out using the manufacturer's instructions.
  • FcyRs The low affinity Fc gamma receptors (FcyRs) such as FcyRlla, FcyRllb, and FcyRllla bind monovalently to the antibody Fc region. Monovalent binding does not activate cells expressing these FcyRs; however, cell-to-cell contact leads to receptor cross-linking and clustering on the cell surface, leading to activation.
  • FcyRllla (CD16) expressed on natural killer (NK) cells
  • target cell killing is mediated by receptor cross-linking and cellular activation, which occurs when the effector cell engages the target cell in a highly avid format (Bowles & Weiner, 2005, J Immunol Methods 304:88-99, Marquez et al., 2010, Cell Immunol 264[1 ]:86-92; expressly incorporated by reference).
  • FcyRllla co-targeting is a particularly good application of the mAb-Fv format, which is capable of engaging target antigen bivalently while co-engaging FcyRllla monovalently.
  • mAb-Fv and mAb-Fab immunoglobulins that co-target FcyRllla were constructed using the variable region of the anti- human FcyRllla antibody 3G8 as Fv-2.
  • a variety of constructs were generated that target various primary antigens (antigen-1 ), including the solid tumor antigens Her2 and EGFR, and the myeloma antigen HM1 .24.
  • Antibody variable regions targeting these antigens were obtained from the anti-Her2 antibody trastuzumab (Carter et al., 1992, Proc Natl Acad Sci USA 89:4285-4289), a humanized version of the anti-EGFR antibody C225 (USSN1 1/004,054), and a humanized version of the anti-HM1.24 antibody HM1 .24 (USSN1 1/857,310).
  • the different constructs incorporated a variety of linker compositions and lengths, and a number of them incorporated hetero-Fc variants that favor heterodimerization.
  • trastuzumab examples include an lgG1 version of trastuzumab, as well as variants with ablated (G236R/L328R) and enhanced (S239D/I332E) binding to FcyR's, including FcyRllla.
  • G236R/L328R is an Fc knockout variant that ablates binding to all FcyRs and complement, and ablates FcyR- and complement- mediated effector function.
  • S239D/I332E enhances affinity to activating FcyRs, and improves FcyR- mediated effector function.
  • Homo Homodimeric native IgG Fc region.
  • Hetero TF-HA Y349T/T394F and
  • d Linker 1 SSDKTHTSPPSPGGGGSGGGG (SEQ ID NO. 43)
  • Linker 2 SSDKTHTSPPSPGGGGSGGGGSGGGGSGGGG (SEQ ID NO. 44)
  • the immunoglobulins in Table 3 were constructed using a combination of gene synthesis (Blue Heron Biotechnology, Bothell, WA), PCR subcloning, quick-change mutagenesis, and other standard molecular biology methods. All DNA was constructed in the context of the expression vector pTT5 (Durocher et al., 2002, Nucleic acids research 30: E9) using standard molecular biology techniques. Heavy and light chains for each
  • immunoglobulin were cotransfected into HEK293E cells (Durocher et al., 2002, Nucleic Acids Research 30: E9) for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography.
  • the immunoglobulins were analyzed using size-exclusion chromatography (SEC) to measure their size (more correctly, their hydrodynamic volume) and determine the nativelike behavior of the purified samples.
  • SEC size-exclusion chromatography
  • HPLC High- performance liquid chromatography
  • the 9516 anti-Her2 x anti-FcyRllla (aHer2 x aFcyRllla) mAb-Fv was constructed in the context of an IgGI comprising the knockout Fc variant (Fc KO) G236R/L328R in order to engineer selective FcyRllla binding. In other words, binding of the Fc region to all of the FcyRs and complement was ablated, and then only FcyRllla binding was built back as antigen-2 in the mAb-Fv format. Binding of 9516 and control antibodies (Native IgGI and Fc KO) to the relevant human FcyR's was tested using surface plasmon resonance (Biacore®).
  • Binding measurements were performed using a Biacore® 3000 instrument (Biacore®). Antibodies were captured onto an immobilized Protein A (Pierce) CM5 biosensor chip (Biacore®), generated using a standard primary amine coupling protocol. All measurements were performed in HBS-EP (0.01 M HEPES pH 7.4, 0.15 M NaCI, 3 mM EDTA, 0.005% v/v surfactant P20), and glycine buffer (10 mM glycine-HCI, pH 1.5) was used for the Protein A surface regeneration. All antibodies (50 nM in HBS-EP) were immobilized on the Protein A surface. Fc receptors were injected over antibody bound surface, followed by a dissociation phase.
  • the Protein A surface was regenerated by injecting glycine buffer (pH 1.5). Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer). The results in Figure 12 show that while native lgG1 binds to all of the FcyRs, the Fc KO variant is ablated for binding to all of the FcyRs.
  • the 9516 aHer2 x aFcyRNIa mAb-Fv is ablated for binding to FcyRI, FcyRlla, and FcyRI lb; however, in contrast to the knockout variant antibody, 9516 has strong affinity for FcyRNIa, due to the engineered Fv-2 binding site.
  • the kinetic off-rate of dissociation (k off ) for FcyRNIa binding is slower than that of native lgG1 , a consequence of the greater affinity of the 3G8 variable region for the receptor relative to the lgG1 Fc region.
  • the neonatal Fc receptor protects IgG from degradation and is responsible in part for the long half-life (-21 days for lgG1 ) of antibodies in circulation.
  • FcRn binds at a site on the Fc region separate from the binding site for FcyRs, and the Fc KO variant
  • G236R/L328R does not impact FcRn affinity. Binding to FcRn by the 9516 mAb-Fv and control antibodies was tested using a Biacore method similar to above, except that a Her2 antigen capture chip format was used (the protein A binding site on Fc overlaps with the site for FcRn). As can be seen in Figure 13, binding of the mAb-Fv to FcRn was uncompromised relative to native lgG1 and the Fc KO variant.
  • Biacore® Affinity of the mAb-Fv for FcyRNIa was measured using Biacore®.
  • the protein A capture format was used as described above, except that sensorgrams were obtained for a concentration series of receptor analtye in order to calculate an accurate affinity.
  • Figure 14 shows Biacore® sensorgrams depicting the binding of various concentrations of the V158 isoform of FcyRNIa to protein A-captured 9516 mAb-Fv. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate
  • a cytotoxicity assay was carried out to test the capacity of the aHer2 x aFcyRl lla mAb-Fv to mediate cellular killing against Her2-expressing cells.
  • a native lgG1 version of the anti-Her2 antibody trastuzumab was run as a control, and an Fc variant version
  • S239D/I332E with enhanced FcyRl l la binding was also tested as a comparator.
  • RSV respiratory syncytial virus
  • Human peripheral blood mononuclear cells (PBMC) were purified from leukopacks using a FicollTM gradient.
  • Her2 + SK-BR-3 breast carcinoma cells were seeded into 96-well plates and opsonized with mAb-Fv or control/comparator antibodies antibodies at the indicated concentrations. Triton X-100 and PBMC alone were run as controls.
  • Effector cells were added at 50: 1 PBMC/target cells, and plates were incubated at 37 °C for 4 h.
  • Cell lysis was measured by lactate dehydrogenase release using the Cytotox-ONETM Homogeneous Membrane Integrity Assay (Promega).
  • Cells were incubated with lactate dehydrogenase reaction mixture for 10 min., and fluorescence was measured using a Wallac Victor2 fluorometer (Perkin-Elmer). Fluorescence was normalized to minimal (PBMC alone) and maximal (Triton) lysis, and fit to a sigmoidal dose-response. The results are shown in Figure 18.
  • the aHer2 x aFcyRll la mAb-Fv mediated a level of lysis substantially enhanced relative to native lgG1 trastuzumab, and comparable to the Fc-enhanced variant S239D/I332E.
  • the results confirm the high affinity FcyRllla (antigen-2) binding mediated by novel engineered Fv-2 binding site, and demonstrate the capacity of the mAb-Fv format for enabling selective and optimal functions.
  • the inhibitory receptor FcyRllb downregulates cellular activation of B cells, monocytes, mast cells, and other immune cell types. On B cells, inhibitory activity is elicited only when FcyRllb rengages into an immune complex with the cell surface B-cell receptor (BCR), a mechanism that is mediated by immune complexation of soluble IgG's with the same antigen that is recognized by the BCR (Heyman 2003, Immunol Lett 88[2]:157-161 ; Smith and Clatworthy, 2010, Nature Reviews Immunology 10:328-343; expressly
  • mAb-Fv immunoglobulins that co-target FcyRllb were constructed using as Fv-2 the variable region of 2B6, an antibody selective for the inhibitory receptor FcyRllb as Fv-2 (Veri et al., 2007, Immunology 121 :392-404).
  • Targeting of CD19 antigen by Fv-1 utilized a humanized affinity matured version of the anti-CD19 antibody 4G7 described previously (USSN 12/156,183).
  • the different constructs incorporated a gly-ser repeat linker, and knockout Fc variants in order to engineer selectivity to FcyRllb relative to other Fc receptors.
  • the mAb-Fv constructs utilized either native CH3 domains (ID 9506) or hetero-Fc variants ID 9547).
  • the anti-FcyRllb (otFcyRllb) mAb-Fv and constructs are listed in Table 4 along with a description of their relevant components. Amino acid sequences of these immunoglobulins are listed in Figure 19. Also included in Table 4 is a control antibody (ID 6187) containing the knockout variant used in these experiments.
  • G236R/L328R is an Fc knockout variant that ablates binding to all FcyRs and complement, and ablates FcyR- and complement- mediated effector function.
  • Homo Homodimeric native IgG Fc region.
  • Hetero TF-HA Y349T/T394F and
  • the immunoglobulins in Table 4 were constructed as described above in the pTT5 expression vector. Heavy and light chains for each immunoglobulin were cotransfected into HEK293E cells for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography, as described above.
  • the 9547 construct was identical 9506 except that it contained the Fc variants Y349T/T394F on one heavy chain and S364H/F405A on the other heavy chain to favor heterodimerization. Multiple bands were observed for 9547 in the SDS-PAGE gels. Bands were observed approximately at the expected molecular weights of the light chain and two heavy chains, and under non-reducing conditions a band was observed at the expected molecular weight of the tetrameric immunoglobulin. Assembly of the mAb-Fv was supported by the presence of an SEC peak that corresponded to the approximate molecular weight of the tetrameric mAb-Fv heterodimer species (data not shown).
  • the size of the additional band in the non-reducing gel suggested a significant population of a monomeric heavy chain / light chain pair (-90 KDa).
  • heavy chain monomers complexed with light chain
  • this result appears to be due to differential expression between the two heavy chains.
  • We suspect the poor expression of one of the heavy chains relative to the other is due to a glycosylation site in the 2B6 VL region (USSN12/349,876). Regardless, the results from the gels and SEC indicate a significant population of the designed heterodimeric mAb-Fv species.
  • binding to of the mAb-Fv is selective for FcyRllb, i.e. it binds the inhibitory receptor FcyRllb with high affinity but does not bind any of the activating FcyRs.
  • Intracellular free calcium concentration [Ca 2+ ] was measured by flow cytometry using a Fluo-4 NW calcium assay (Molecular Probes, Eugene, OR).
  • PBMCs were resuspended in calcium assay buffer and pre-loaded with Fluo-4 dye for 30 min at room temperature. After incubation with immunoglobulin of interest, cells were stimulated by addition of 10 ⁇ g/ml of anti-CD79b antibody.
  • Calcium flux kinetics was recorded using a FACSCanto II flow cytometer and data were analyzed using FlowJo software (Tree Star, Ashland, OR). The data are plotted as the change of Mean Fluo-4 Intensity (MFI) over time. The results are shown in Figure 22.
  • T-cell receptor TCR
  • mAb-Fv immunoglobulins that co-target CD3 were constructed using as the Fv-2 variable region a humanized version of the anti-CD3 antibody OKT3.
  • Fv-1 regions targeted the antigens Her2, HM1 .24, and CD19 using the variable regions described above.
  • a variety of constructs were generated co-targeting these primary antigens bivalently and CD3 monovalently that incorporated different linker compositions and lengths, hetero-Fc variants that favor heterodimerization, and knockout Fc variants to ablate binding to FcyRs and complement. Constructs also incorporated His- and Flag- tags at the C-termini as potential detection/purification tags.
  • the aCD3 mAb-Fv constructs are listed in Table 5 along with a description of their relevant components. Amino acid sequences of these immunoglobulins are listed in Figure 23.
  • G236R/L328R is an Fc knockout variant that ablates binding to all FcyRs and complement, and ablates FcyR- and complement- mediated effector function.
  • Homo Homodimeric native IgG Fc region.
  • Hetero TF-HA Y349T/T394F and
  • Linker 4 SSDKTHTSPPSPSG (SEQ ID NO. 46)
  • the immunoglobulins in Table 5 were constructed as described above in the pTT5 expression vector. Heavy and light chains for each immunoglobulin were cotransfected into HEK293E cells for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography, as described above. [276] The proteins were visualized electrophoretically using the Agilent Bioanalyzer as described above. Representative gels are shown in Figure 24. As expected, under reducing conditions three bands were observed at the approximate calculated molecular weights, corresponding to the light chain (-25 KDa) and two heavy chains (-65 KDa) for each mAb- Fv.
  • Affinity of the 9509 aHer2 x aCD3 mAb-Fv for CD3 + T cells was measured using a competition experiment with labeled OKT3 antibody.
  • 9509 mAb-Fv at various concentrations was pre-incubated with -0.8 million PBMC on ice for 1 hour.
  • Commercial FITC-labeled OKT3 was added to 50 ng/ml for 45 minutes.
  • the binding buffer was 3% FBS/PBS, and samples were washed twice before fixing with 1 % Paraformaldehyde/PBS.
  • aHer2 x aCD3 mAb-Fv's utilized purified T cells as effectors.
  • T cells were purified using a T Cell Enrichment Kit from Stemcell Technologies (Vancouver, Canada).
  • the 9509 aHer2 x aCD3 mAb-Fv was tested against Her2 + SK-BR-3 breast carcinoma cells at an effector to target ratio of 200,000 T cells to 10,000 target cells. Incubation time was 24 hrs.
  • the 9551 aHM1 .24 x aCD3 mAb-Fv was used as a negative control.
  • the results in Figure 27 show strong CD3-mediated cytotoxicity against the Her2 + target cells mediated by the Her2-directed mAb-Fv, while the HM1 .24-directed mAb-Fv showed no killing activity.
  • the 9344 aHer2 x aCD3 mAb-Fv was tested against Her2 + SKOV3 ovarian carcinoma cells at an effector to target ratio of 250,000 T cells to 10,000 target cells. Incubation time was 24 hrs.
  • the aHer2 lgG1 WT antibody trastuzumab was used as a negative control.
  • the results in Figure 28 show strong CD3-mediated killing against the target cells by the mAb-Fv.
  • the WT lgG1 anti-Her2 antibody showed no capacity to mediate cytotoxic activity by the isolated T cells.
  • HM1 .24 + RPMI 8224 myeloma cells were used as targets (seeded at 10,000 per well), and 500,000 PBMCs were added as effectors at a 50:1 effector/target cell ratio. Incubation time for this assay was 23 hrs.
  • the results in Figure 29 show that the aHM1 .24 x aCD3 mAb-Fv mediated strong cytotoxic activity against the tumor cells.
  • the 6166 Fc knockout anti- HMI .24 antibody was completely devoid of activity, consistent with it's lack of FcyR binding, and of course its inability as a native antibody to co-engage CD3.
  • the 5627 WT lgG1 anti- HMI .24 antibody mediated very weak ADCC activity, if any at all, presumably due to FcyRllla-mediated killing by NK cells in the PBMC mixture.
  • the dramatic CD3-mediated activity of the mAb-Fv relative to the FcyR-mediated activity of the normal lgG1 antibody targeting the same antigen demonstrates the potential of the novel formats herein for enabling powerful functional activities.

Abstract

Immunoglobulin compositions that simultaneously co-engage antigens, where one of the antigens is bound bivalently and the other antigen is bound monovalently. The novel immunoglobulins described preferably utilize heterodimeric Fc regions.

Description

COMPOSITIONS AND METHODS FOR SIMULTANEOUS BIVALENT AND MONOVALENT
CO-ENGAGEMENT OF ANTIGENS
RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Patent Application Serial No. 61/239,316, filed September 2, 2009 under 35 U.S.C. §1 19(e) and incorporates the same in its entirety herein.
TECHNICAL FIELD
[1] The present disclosure provides novel antibody analog compositions that
simultaneously co-engage antigens, where one of the antigens is bound bivalently and the other antigen is bound monovalently. The novel antibody analogs described preferably utilize heterodimeric Fc regions. Methods of using the novel antibody analog compositions, particularly for therapeutic purposes, are also described herein.
BACKGROUND
[2] Antibody-based therapeutics have been used succesfully to treat a variety of diseases, including cancer and autoimmune/inflammatory disorders. Yet improvements to this class of drugs are still needed, particularly with respect to enhancing their clinical efficacy. One avenue being explored is the engineering of additional and novel antigen binding sites into antibody-based drugs such that a single immunoglobulin molecule co- engages two different antigens. Such non-native or alternate antibody formats that engage two different antigens are often referred to as bispecifics. Because the considerable diversity of the antibody variable region (Fv) makes it possible to produce an Fv that recognizes virtually any molecule, the typical approach to bispecific generation is the introduction of new variable regions into the antibody.
[3] A number of alternate antibody formats have been explored for bispecific targeting (Chames & Baty, 2009, mAbs 1 [6]:1-9; Holliger & Hudson, 2005, Nature Biotechnology 23[9]:1 126-1 136; expressly incorporated herein by reference). Initially, bispecific antibodies were made by fusing two cell lines that each produced a single monoclonal antibody (Milstein et al., 1983, Nature 305:537-540). Although the resulting hybrid hybridoma or quadroma did produce bispecific antibodies, they were only a minor population, and extensive purification was required to isolate the desired antibody. An engineering solution to this was the use of antibody fragments to make bispecifics. Because such fragments lack the complex quaternary structure of a full length antibody, variable light and heavy chains can be linked in single genetic constructs. Antibody fragments of many different forms have been generated, including diabodies, single chain diabodies, tandem scFv's, and Fab2 bispecifics (Chames & Baty, 2009, mAbs 1 [6]:1 -9; Holliger & Hudson, 2005, Nature
Biotechnology 23[9]:1 126-1 136; expressly incorporated herein by reference). While these formats can be expressed at high levels in bacteria and may have favorable penetration benefits due to their small size, they clear rapidly in vivo and can present manufacturing obstacles related to their production and stability. A principal cause of these drawbacks is that antibody fragments typically lack the constant region of the antibody with its associated functional properties, including larger size, high stability, and binding to various Fc receptors and ligands that maintain long half-life in serum (i.e. the neonatal Fc receptor FcRn) or serve as binding sites for purification (i.e. protein A and protein G).
[4] More recent work has attempted to address the shortcomings of fragment-based bispecifics by engineering dual binding into full length antibody -like formats (Wu et al., 2007, Nature Biotechnology 25[1 1]:1290-1297; USSN12/477.71 1 ; Michaelson et al., 2009, mAbs 1 [2]:128-141 ; PCT/US2008/074693; Zuo et al., 2000, Protein Engineering 13[5]:361 -367; USSN09/865,198; Shen et al., 2006, J Biol Chem 281 [16]: 10706-10714; Lu et al., 2005, J Biol Chem 280[20]: 19665-19672; PCT/US2005/025472; expressly incorporated herein by reference). These formats overcome some of the obstacles of the antibody fragment bispecifics, principally because they contain an Fc region. One significant drawback of these formats is that, because they build new antigen binding sites on top of the homodimeric constant chains, binding to the new antigen is always bivalent.
[5] For many antigens that are attractive as co-targets in a therapeutic bispecific format, the desired binding is monovalent rather than bivalent. For many immune receptors, cellular activation is accomplished by cross-linking of a monovalent binding interaction. The mechanism of cross-linking is typically mediated by antibody/antigen immune complexes, or via effector cell to target cell engagement. For example, the low affinity Fc gamma receptors (FcyRs) such as FcyRlla, FcyRllb, and FcyRllla bind monovalently to the antibody Fc region. Monovalent binding does not activate cells expressing these FcyRs; however, upon immune complexation or cell-to-cell contact, receptors are cross-linked and clustered on the cell surface, leading to activation. For receptors responsible for mediating cellular killing, for example FcyRllla on natural killer (NK) cells, receptor cross-linking and cellular activation occurs when the effector cell engages the target cell in a highly avid format (Bowles & Weiner, 2005, J Immunol Methods 304:88-99, expressly incorporated by reference).. Similarly, on B cells the inhibitory receptor FcyRllb downregulates B cell activation only when it engages into an immune complex with the cell surface B-cell receptor (BCR), a
mechanism that is mediated by immune complexation of soluble IgG's with the same antigen that is recognized by the BCR (Heyman 2003, Immunol Lett 88[2]:157-161 ; Smith and Clatworthy, 2010, Nature Reviews Immunology 10:328-343; expressly incorporated by reference). As another example, CD3 activation of T-cells occurs only when its associated T- cell receptor (TCR) engages antigen-loaded MHC on antigen presenting cells in a highly avid cell-to-cell synapse (Kuhns et al., 2006, Immunity 24:133-139). Indeed nonspecific bivalent cross-linking of CD3 using an anti-CD3 antibody elicits a cytokine storm and toxicity (Perruche et al., 2009, J Immunol 183[2]:953-61 ; Chatenoud & Bluestone, 2007, Nature Reviews Immunology 7:622-632; expressly incorporated by reference). Thus for practical clinical use, the preferred mode of CD3 co-engagement for redirected killing of targets cells is monovalent binding that results in activation only upon engagement with the co-engaged target.
[6] Thus while bispecifics generated from antibody fragments suffer biophysical and pharmacokinetic hurdles, a drawback of those built with full length antibody -like formats is that they engage co-target antigens multivalently in the absence of the primary target antigen, leading to nonspecific activation and potentially toxicity. The present invention solves this problem by introducing a novel set of bispecific formats that enable the simultaneous bivalent and monovalent co-engagement of distinct target antigens.
SUMMARY
[7] The present disclosure provides novel antibody analog compositions that co-engage a first and second antigen.
BRIEF DESCRIPTION OF THE DRAWINGS
[8] Figure 1. Heterodimeric Fc region proof-of-concept system. DNA constructs encoding two different immunoglobulin polypeptides were designed: (i) scFv-Fc (VH-linker-VK-Hinge- CH2-CH3') and (ii) empty-Fc (Hinge-CH2-CH3"). As shown, three possible dimers can form from cotransfection and expression of these two polypeptides, each composed of a different number of immunoglobulin (Ig) domains and thus different molecular weights. The two different CH3 domains, CH3' (prime) and CH3" (double prime), can be designed to promote heterodimer formation and discourage homodimer formation.
[9] Figure 2. Amino acid sequences of heterodimer constructs and native lgG1 and lgG2 constant heavy chains (SEQ ID NOs. 1-4). [10] Figure 3. Electrophoretic visualization of protein species generated by simultaneous expression of empty-Fc and scFv-Fc polypeptides (each with wild-type lgG1 Fc sequence) using Agilent 2100 Bioanalyzer microfluidics-based platform. The molecular weights of the proteins in the ladder lane (designated L) are shown on the left in kilodaltons (KDa). The other lane assignments are as follows: Lanes 1-5: reducing environment, cotransfection DNA ratios of empty-Fc:scFv-Fc 1 :1 , 1 :2, 1 :4, 2:1 , 4:1 ; Lanes 6-10: non-reducing
environment, empty-Fc:scFv-Fc DNA ratios 1 :1 , 1 :2, 1 :4, 2:1 , 4:1 . In lanes 1 -5, the top band (-70 KDa) is scFv-Fc monomer, and the bottom band (-38 kDa) is empty-Fc monomer. In lanes 6-10, the top band (-134 KDa) is scFv-Fc homodimer, the middle band (-105 KDa) is scFc-Fv/empty-Fc heterodimer, and the bottom band (-72 KDa) is empty-Fc homodimer.
[11] Figure 4. Electrophoretic visualization of protein species generated by simultaneous expression of variant empty-Fc and variant scFv-Fc (cotransfection DNA ratio of empty- Fc:scFv-Fc = 2:3) using the Agilent Bioanalyzer. Environment is non-reducing. The molecular weights of the proteins in the ladder lane (designated L) are shown on the left in kilodaltons (KDa). The other lane assignments at the top are as given in Figure 5, column 1 .
[12] Figure 5. Protein species observed upon simultaneous expression of variant empty- Fc and variant scFv-Fc (cotransfection DNA ratio of empty-Fc:scFv-Fc = 2:3). Relative concentrations were determined using the Agilent Bioanalyzer (see Figure 4 for gel results). Empty-Fc refers to substitutions in the Empty-Fc chain and scFv-Fc refers to substitutions in the scFv-Fc chain. The last five columns list for each variant the molar percentage of Empty- Fc monomer (Empty-M), Empty-Fc dimer (Empty-D), Heterodimer (Hetero), scFv-Fc dimer (scFv-D), and scFv-Fc monomer (scFv-M).
[13] Figure 6. Protein species by molar % from simultaneous expression of variant empty- Fc and variant scFv-Fc. Empty-Fc refers to substitutions in the Empty-Fc chain and scFv-Fc refers to substitutions in the scFv-Fc chain. The last five columns list for each variant the molar percent of Empty-Fc monomer (Empty-M), Empty-Fc dimer (Empty-D), Heterodimer (Hetero), scFv-Fc dimer (scFv-D), and scFv-Fc monomer (scFv-M).
[14] Figure 7. Protein species by mass % from simultaneous expression of variant empty- Fc and variant scFv-Fc. Empty-Fc refers to substitutions in the Empty-Fc chain and scFv-Fc refers to substitutions in the scFv-Fc chain. The last five columns list for each variant the mass percent of Empty-Fc monomer (Empty-M), Empty-Fc dimer (Empty-D), Heterodimer (Hetero), scFv-Fc dimer (scFv-D), and scFv-Fc monomer (scFv-M).
[15] Figure 8. Illustration of mAb-Fv and mAb-Fab immunoglobulin formats. A native lgG1 antibody is included for reference. Although mAb-Fv and mAb-Fab immunoglobulins can be constructed with homodimeric CH3 domains, preferably they utilize heterodimeric CH3 domains, as depicted. Bivalent binding to antigen-1 (Ag-1 ) is mediated by the N-terminal VH/VL pairs (Fv-1 ), while monovalent binding to antigen-2 (Ag-2) is mediated by the C- terminal VH/VL pair (Fv-2). Also shown are the Fab-Fv and Fab-Fab analogs that bind both antigen-1 and antigen-2 monovalently.
[16] Figure 9. Sequences of mAb-Fv constructs with Fv-2 targeting FcYRIIIa. LC refers to light chain, and HC1 and HC2 refer to the two heavy chains that make up each mAb-Fv. Identification (ID) numbers correspond to those in Table 3 and the results. 9064 is a mAb- Fab, all other constructs are mAb-Fv's. For each construct the linker separating the Fc region with Fv-2 is underlined, and Fv-2 and any C-terminal linkers are shown in italics. In addition, the histidine tags in enumerated sequences are optional, and generally not included in therapeutic compositions (SEQ ID NOs. 5-21 ).
[17] Figure 10. Electrophoretic visualization of mAb-Fvs and mAb-Fab with Fv-2 targeting FcYRIIIa using the Agilent Bioanalyzer. Environment is non-reducing ("nr") or reducing ("red") as labeled. The molecular weights of the proteins in the ladder lane (L) are shown on the left in kilodaltons (KDa).
[18] Figure 1 1 . Size exclusion chromatograms for aFcYRIIIa co-engaging mAb-Fv's 9516, 9517, and 9518.
[19] Figure 12. Biacore® sensorgrams depicting the binding of various FCYRS to mAb-Fv and antibodies captured by a protein A chip. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer).
[20] Figure 13. Biacore® sensorgrams depicting the binding of FcRn to mAb-Fv and antibodies captured by a HER2 chip. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer).
[21] Figure 14. Biacore® sensorgrams depicting the binding of various concentrations of FcYRIIIa(V158 isoform) to protein A-captured 9516 mAb-Fv. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer). The Kd of 1 1 1 nM was calculated by global fitting of binding data with a 1 :1 Langmuir binding model using BIAevaluation software.
[22] Figure 15. Biacore® sensorgrams depicting the simultaneous co-engagement of Her2 and Fc Rllla by mAb-Fv. 9516 (aHER2 x aFcYRIIIa) was captured to a HER2 chip followed by binding of FcYRII Ia(158V). Data were processed by zeroing time and response before the injection of 9516.
[23] Figure 16. Biacore® sensorgrams depicting the binding of various concentrations of FcYRI I Ia(158V) to HM1 .24-captured 9518. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer). The Kd of 96 nM was calculated by global fitting of binding data with a 1 : 1 Langmuir binding model using
BIAevaluation software.
[24] Figure 17. Biacore sensorgrams depicting the capture of 9518 (αΗΜ1 .24 x aFcYRII Ia) by a HM 1 .24 chip followed by dual binding of FcYRI I Ia(158V). Data were processed by zeroing time and response before the injection of 9518.
[25] Figure 18. Cytotoxicity of various mAb-Fvs and mAbs against SK-BR-3 cells using human PBMCs. FcYRI IIa-dependent cytotoxicity was measured at multiple mAb-Fv or mAb concentrations by lactate dehydrogenase release (mean ± SE of triplicate wells).
[26] Figure 19. Sequences of mAb-Fv constructs with Fv-2 targeting FcYRI Ib. In addition, the histidine tags in enumerated sequences are optional, and generally not included in therapeutic compositions (SEQ I D NOs. 22-26).
[27] Figure 20. Electrophoretic visualization of mAb-Fvs with Fv-2 targeting FcYRI Ib using the Agilent Bioanalyzer. Environment is non-reducing ("nr") or reducing ("red") as labeled. The molecular weights of the proteins in the ladder lane (L) are shown on the left in kilodaltons (KDa).
[28] Figure 21 . Biacore® sensorgrams depicting the binding of various concentrations of FcYRI Ib to protein A-captured 9547 mAb-Fv. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer). The Kd of 44 nM was calculated by global fitting of binding data with a 1 : 1 Langmuir binding model using
BIAevaluation software.
[29] Figure 22. Calcium response of CD40+ PBMCs after incubation with various concentrations of 9547 (aCD19 x aFcYRI Ib) or 6187 (aCD19 FcKO). Calcium mobilization was induced with 10 [ig/m\ anti-CD79b (G236R/L328R) BCR-activating antibody.
Intracellular free calcium concentration was measured by flow cytometry using a Fluo-4 NW calcium assay. Mean fluorescence intensity was based on a 30 second running average. [30] Figure 23. Sequences of mAb-Fv constructs with Fv-2 targeting CD3. In addition, the histidine tags in enumerated sequences are optional, and generally not included in therapeutic compositions (SEQ ID NOs. 27-42).
[31] Figure 24. Electrophoretic visualization of mAb-Fvs with Fv-2 targeting CD3 using the Agilent Bioanalyzer. Environment is non-reducing ("nr") or reducing ("red") as labeled. The molecular weights of the proteins in the ladder lane (L) are shown on the left in kilodaltons (KDa).
[32] Figure 25. Size exclusion chromatograms for aCD3 co-engaging mAb-Fvs.
[33] Figure 26. Competion of unlabeled 9509 aHER2 x aCD3 mAb-Fv with FITC-labeled full-length OKT3 mAb for binding to CD3+ T cells in PBMCs.
[34] Figure 27. Cytotoxicity of mAb-Fvs against Her2+ SK-BR-3 cells using T cells isolated from human PBMCs. CD3-dependent cytotoxicity was measured at multiple mAb-Fv concentrations by lactate dehydrogenase release (mean ± SE of triplicate wells).
[35] Figure 28. Cytotoxicity of mAb-Fvs against Her2+ SKOV3 cells using T cells isolated from human PBMCs. CD3-dependent cytotoxicity was measured at multiple mAb-Fv concentrations by lactate dehydrogenase release (mean ± SE of triplicate wells).
[36] Figure 29. Cytotoxicity of various mAb-Fvs and mAbs against HM1 .24+ RPMI 8224 cells using human PBMCs. CD3-dependent cytotoxicity was measured at multiple mAb-Fv or mAb concentrations by lactate dehydrogenase release (mean ± SE of triplicate wells).
DETAILED DESCRIPTION OF THE INVENTION
[37] Overview
[38] The present invention is directed to a variety of antibodies (generally referred to herein as "antibody analogs", as further described below) that co-engage at least two different antigens in novel ways. The two antigens of the invention are generally referred to as antigen-1 and antigen-2 (with some embodiments, described below, including antigen-3 as a third antigen). In general, there are two different types of antibody analogs that allow for co-engagement mechanisms, one that utilizes three antigen binding domains (e.g. one antigen is bound bivalently and the other is bound monovalently, although as is further described below, there can also be three different antigens that are bound or a single antigen), and one that relies on two antigen binding domains (e.g. each antigen is bound monovalently).
[39] In one embodiment, antibody analogs are made that have three antigen binding domains. This embodiment can be exemplified in a variety of ways. In an embodiment of particular use, this is done by using a "traditional" antibody structure (two heavy chains with associated light chains that form antigen binding domains) that includes an additional antigen binding domain at the "opposite end" of the antibody. That is, in some
embodiments, all three of the antigen binding domains of the antibody analog are antibody variable regions, comprising a variable heavy and variable light chain.
[40] Generally, in this embodiment, two of the antigen binding domains bind to antigen-1 , e.g. bivalently, and the third antigen binding domain binds monovalently to antigen-2. As discussed below, there are particular therapeutic embodiments for which monovalent binding can be preferred, to avoid cross-linking and/or activation of certain pathways.
[41] As outlined in Figure 8, this "trimeric binding domain" embodiment can be
accomplished using two different heavy chains; that is, the antibody analog is
"heterodimeric", as defined below. These heterodimeric heavy chains differ from one another in one or more ways. In general, one of the heavy chains has a C-terminal fusion (e.g. fused to the C-terminus of the CH3 domain of the first heavy chain) that comprises an "extra" (sometimes referred to herein as "exogenous") variable heavy domain. The other heavy chain also has a C-terminal fusion that comprises an exogenous variable light domain. Together the two "extra" variable domains form an antigen binding domain. Thus, in addition to the two "traditional" binding domains, these antibody analogs have a third binding domain.
[42] Optionally, but preferred in some embodiments, the two heavy chains also differ from each other by the presence of amino acid substitutions in the Fc domain that favors heterodimeric formation over homodimers. That is, by changing amino acids in each heavy chain, different heavy chains are more likely to associate to form the antibody analog structure than to form homodimers with the same Fc amino acid sequences.
[43] In another embodiment, antibody analogs are made that have two different antigen binding domains (although as described below, the antigens could be identical). While these antibody analogs share some functional similarity to bispecific antibodies and some antibody fragment constructs, the bispecific antibody analogs of the present invention are easier to make and use than traditional bispecific antibodies.
[44] In this embodiment, the "dimeric binding domain" antibody analog also has components of traditional antibodies. The first heavy chain is identical to a "traditional" antibody construct, in that in contains the variable heavy and light regions, with the exception that it contains a C-terminal fusion at the CH3 domain of an exogenous variable domain (either heavy or light). The second heavy chain comprises the Fc region of the heavy chain and an N-terminal variable light chain instead of a N-terminal variable heavy chain, such that the N-terminal portion of the analog forms a first antigen binding site (e.g. binds to antigen- 1 ). The second heavy chain also contains a C-terminal fusion to the CH3 domain of an exogeneous variable domain, either heavy or light, such that one chain contains the exogenous heavy chain and the other the exogenous light chain. This general scheme is shown in Figure 8.
[45] In some embodiments of the invention, the antigen binding regions of the antibody analog are antibody variable regions. In these embodiments, binding to antigens is mediated by variable regions, also referred to as Fv regions, each comprising a VH domain and a VL domain. The Fv region that binds antigen-1 is referred to as Fv-1 , while the Fv region that binds antigen-2 is referred to as Fv-2.
[46] In some preferred embodiments of the invention, the antibody analog is a novel antibody format referred to as mAb-Fv or mAb-Fab. As described herein, mAb-Fv comprises an IgG antibody with a C-terminal Fv fusion. mAb-Fab comprises an IgG antibody with a C- terminal Fab fusion. The N-terminal Fab regions of the mAb-Fv and mAb-Fab contain Fv-1 . Because there are two such Fv-1 regions, the mAb-Fv and mAb-Fab bind bivalently to antigen-1 . That is, one immunoglublin binds two of antigen-1 at the same time. In contrast, the C-terminal Fv region of the mAb-Fv or C-terminal Fab region of the mAb-Fab contain Fv- 2, which is the binding site for antigen-2. Because there is only one such Fv-2 region, the mAb-Fv and mAb-Fab bind monovalently to antigen-2. That is, one immunoglobulin binds only one of antigen-2. The mAb-Fv and mAb-Fab immunoglobulins thus contain three variable regions, two of which are Fv-1 and one of which is Fv-2. The mAb-Fab differs from the mAb-Fv in that the former contains CH and CL constant domains C-terminal to the Fv-2 VH and VL domains respectively.
[47] As described herein, the mAb-Fv and mAb-Fab immunoglobulins comprise three distinct immunoglobulin chains. These chains include an antibody light chain and two antibody heavy chains. One of said heavy chains comprises a VH or VH-CH 1 at its C- terminus, whereas the other of said heavy chains comprises a VL or VL-CL at its C-terminus.
[48] Also described herein are analogues of the mAb-Fv and mAb-Fab that bind both antigen-1 and antigen-2 monovalently. These immunoglobulins are referred to as Fab-Fv and Fab-Fab. The Fab-Fv and Fab-Fab immunoglobulins comprise an N-terminal Fab region that contains a single binding site for antigen-1 , and comprise a C-terminal Fv or Fab region that contains a single binding site for antigen-2.
[49] mAb-Fv and mAb-Fab immunoglobulins of the invention may also comprise single domain VHH regions as Fv-1 . That is, the N-terminal binding portion of the mAb-Fv and mAb-Fab may lack a light chain and a CH 1 domain. [50] In some preferred embodiment of the invention, the first and second antigens are distinct. That is, Fv-1 and Fv-2 bind different antigens. However, in some embodiments, antigen-1 and antigen-2 may be the same, i.e. Fv-1 and Fv-2 have specificity for the same antigen.
[51] Also contemplated herein are an immunoadhesin-Fv and immunoadhesin-Fab. In this format, Fv-1 is replaced with a fusion protein, for example a receptor, that binds antigen-1. Thus antigen-1 is bound by a fusion protein, while antigen-2 is bound by Fv-2.
[52] The novel antibody analogs described preferably comprise a heterodimeric Fc region. A heterodimer Fc region, as described herein, is a pair of variant Fc regions that comprise at least one amino acid modification in each Fc region. In a preferred embodiment, said amino acid modifications stabilize a heterodimeric Fc region while destabilizing homodimeric Fc regions. In one embodiment of the invention, said variant Fc regions comprise at least one substitution at a position selected from the group consisting of 349, 351 , 354, 356, 357, 364, 366, 368, 370, 392, 394, 395, 396, 397, 399, 401 , 405, 407, 409, 41 1 , and 439, wherein numbering is according to the EU index as in Kabat. In a preferred embodiment, said variant Fc regions comprise at least one substitution selected from the group consisting of 349A, 349C, 349E, 349I, 349K, 349S, 349T, 349W, 351 E, 351 K, 354C, 356K, 357K, 364C, 364D, 364E, 364F, 364G, 364H, 364R, 364T, 364Y, 366D, 366K, 366S, 366W, 366Y, 368A, 368E, 368K, 368S, 370C, 370D, 370E, 370G, 370R, 370S, 370V, 392D, 392E, 394F, 394S, 394W, 394Y, 395T, 395V, 396T, 397E, 397S, 397T, 399K, 401 K, 405A, 405S, 407T, 407V, 409D, 409E, 41 1 D, 41 1 E, 41 1 K, and 439D.
[53] The present invention describes methods for generating the novel compositions of the invention. The present invention describes purification methods for the immunogloublins herein, particularly methods for separating heterodimeric and homodimeric protein species. Also described are methods of testing the immunogloublins herein, including in vitro and in vivo experiments.
[54] The present invention provides isolated nucleic acids encoding the novel antibody analog compositions described herein. The present invention provides vectors comprising said nucleic acids, optionally, operably linked to control sequences. The present invention provides host cells containing the vectors, and methods for producing and optionally recovering the antibody analog compositions.
[55] The present invention provides compositions comprising antibody analog
polypeptides described herein, and a physiologically or pharmaceutically acceptable carrier or diluent. [56] The present invention contemplates therapeutic and diagnostic uses for the antibody analog polypeptides disclosed herein.
[57]
[58] Definitions
[59] In order that the invention may be more completely understood, several definitions are set forth below. Such definitions are meant to encompass grammatical equivalents.
[60] By "amino acid" and "amino acid identity" as used herein is meant one of the 20 naturally occurring amino acids or any non-natural analogues that may be present at a specific, defined position. Thus "amino acid" as used herein means both naturally occurring and synthetic amino acids. For example, homophenylalanine, citrulline and noreleucine are considered amino acids for the purposes of the invention. "Amino acid" also includes imino acid residues such as proline and hydroxyproline. The side chain may be in either the (R) or the (S) configuration. In the preferred embodiment, the amino acids are in the (S) or L- configuration. If non-naturally occurring side chains are used, non-amino acid substituents may be used, for example to prevent or retard in vivo degradation.
[61]
[62] By "constant region" of an antibody as defined herein is meant the region of the antibody that is encoded by one of the light or heavy chain immunoglobulin constant region genes. By "constant light chain" or "light chain constant region" or "CL" as used herein is meant the region of an antibody encoded by the kappa (CK) or lambda (C ) light chains. The constant light chain typically comprises a single domain, and as defined herein refers to positions 1 08-214 of CK or C , wherein numbering is according to the EU index. By
"constant heavy chain" or "heavy chain constant region" as used herein is meant the region of an antibody encoded by the mu, delta, gamma, alpha, or epsilon genes to define the antibody's isotype as IgM, IgD, IgG, IgA, or IgE, respectively. For full length IgG antibodies, the constant heavy chain, as defined herein, refers to the N-terminus of the CH 1 domain to the C-terminus of the CH3 domain, thus comprising positions 1 1 8-447, wherein numbering is according to the EU index.
[63] By "Fab" or "Fab region" as used herein is meant the polypeptides that comprise the VH, CH 1 , VL, and CL immunoglobulin domains. Fab may refer to this region in isolation, or this region in the context of a full length antibody or antibody fragment.
[64] By "Fc" or "Fc region", as used herein is meant the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain. Thus Fc refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N-terminal to these domains. For IgA and IgM, Fc may include the J chain. For IgG, Fc comprises immunoglobulin domains Cgamma2 and Cgamma3 (Cy2 and Cy3) and the hinge between Cgammal (Cy1 ) and Cgamma2 (Cy2). Although the boundaries of the Fc region may vary, the human IgG heavy chain Fc region is usually defined to comprise residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat. Fc may refer to this region in isolation, or this region in the context of an Fc polypeptide, for example an antibody. By "Fc polypeptide" as used herein is meant a polypeptide that comprises all or part of an Fc region. Fc polypeptides include antibodies, Fc fusions, isolated Fes, and Fc fragments.
[65] By "Fc fusion" or "immunoadhesin" as used herein is meant a protein wherein one or more polypeptides is operably linked to an Fc domain. Fc fusion is herein meant to be synonymous with the terms "immunoadhesin", "Ig fusion", "Ig chimera", and "receptor globulin" as used in the prior art (Huang, 2009, Curr Opin Biotechnology 20:692-699;
Chamow et al., 1996, Trends Biotechnol 14:52-60; Ashkenazi et al., 1997, Curr Opin
Immunol 9:195-200, both hereby entirely incorporated by reference). An Fc fusion combines the Fc region of an immunoglobulin with a fusion partner, which in general may be any protein, polypeptide or small molecule. The role of the non-Fc part of an Fc fusion, i.e., the fusion partner, is to mediate target binding, and thus it is functionally analogous to the variable regions of an antibody. Virtually any protein or small molecule may be linked to Fc to generate an Fc fusion. Protein fusion partners may include, but are not limited to, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain. Small molecule fusion partners may include any therapeutic agent that directs the Fc fusion to a therapeutic target. The role of the latter is to mediate target recognition, and thus it is functionally analogous to the antibody variable region. Because of the structural and functional overlap of Fc fusions with antibodies, the discussion on antibodies in the present disclosure extends also to Fc fusions.
[66] Virtually any protein or small molecule may be linked to Fc to generate an Fc fusion. Protein fusion partners may include, but are not limited to, the variable region of any antibody, the target-binding region of a receptor, an adhesion molecule, a ligand, an enzyme, a cytokine, a chemokine, or some other protein or protein domain. Small molecule fusion partners may include any agent that directs the Fc fusion to a target antigen. Such target antigen may be any molecule, e.g., an extracellular receptor, that is implicated in disease. Fusion partners may be linked to any region of an Fc region, including at the N- or C- termini, or at some residue in-between the termini. In one embodiment, a fusion partner is linked at the N- or C-terminus of the Fc region. [67] By "Fv region" or "variable region" when used herein refers to the minimum fragment of an antibody that retains both antigen-recognition and antigen-binding sites.
[68] By "heterodimeric Fc region" as used herein is meant an Fc region composed of two different heavy constant chains, e,g, having different sequences . As described more fully below, the changes can comprise one or more amino acid changes.
[69] By "immunoglobulin (Iq)" as used herein is meant a protein consisting of one or more polypeptides substantially encoded by immunoglobulin genes. Immunoglobulins include but are not limited to antibodies. Immunoglobulins may have a number of structural forms, including but not limited to full length antibodies, antibody fragments, and individual immunoglobulin domains. By "immunoglobulin (Iq) domain" herein is meant a region of an immunoglobulin that exists as a distinct structural entity as ascertained by one skilled in the art of protein structure. Ig domains typically have a characteristic β-sandwich folding topology. By "immunoglobulin isotype" or "isotype" as used herein is meant any of the subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions. The known human immunoglobulin isotypes are lgG1 , lgG2, lgG3, lgG4, lgA1 , lgA2, IgM, IgD, and IgE.
[70] By "IgG" as used herein is meant a polypeptide belonging to the class of antibodies that are substantially encoded by a recognized immunoglobulin gamma gene. In humans this class comprises lgG1 , lgG2, lgG3, and lgG4. In mice this class comprises lgG1 , lgG2a, lgG2b, lgG3. As described herein and in the references below, IgG molecules also include hybrid IgG molecules, such as hybrids of lgG1/G2 described in U.S. Publication No.
2006/0134150, and IgG molecules that have amino acid substitutions as compared to wild- type IgG molecules. Thus, IgG molecules can include naturally occurring molecules or those with sequence modifications.
[71] By "parent polypeptide", "parent protein", "precursor polypeptide", or "precursor protein" as used herein is meant an unmodified polypeptide that is subsequently modified to generate a variant. Said parent polypeptide may be a naturally occurring polypeptide, or a variant or engineered version of a naturally occurring polypeptide. Parent polypeptide may refer to the polypeptide itself, compositions that comprise the parent polypeptide, or the amino acid sequence that encodes it. Accordingly, by "parent Fc polypeptide" as used herein is meant an Fc polypeptide that is modified to generate a variant, and by "parent antibody" as used herein is meant an antibody that is modified to generate a variant antibody.
[72] By "protein" or "polypeptide" as used herein is meant at least two covalently attached amino acids, which includes proteins, polypeptides, oligopeptides and peptides. The protein may be made up of naturally occurring amino acids and peptide bonds, or synthetic peptidomimetic structures, i.e. "analogs", such as peptoids.
[73] By "position" as used herein is meant a location in the sequence of a protein or nucleic acid. Protein positions may be numbered sequentially, or according to an established format, for example the Kabat index for antibody variable regions or the EU index for antibody constant regions. For example, position 297 is a position in the human antibody lgG1. Corresponding positions are determined as outlined above, generally through alignment with other parent sequences.
[74] By "residue" as used herein is meant a position in a protein and its associated amino acid identity. For example, Asparagine 297 (also referred to as Asn297, also referred to as N297) is a residue in the human antibody lgG1 . In some embodiments it can also refer to nucleic acid bases.
[75] By "target antigen" or "target" or "antigen" as used herein is meant the molecule that is bound specifically by the variable region of an antibody, or by the fusion protein of an Fc fusion. A target antigen may be a protein, carbohydrate, lipid, or other chemical compound. A wide variety of suitable targets are described below.
[76] By "target cell" as used herein is meant a cell that expresses a target antigen.
[77] By "variant polypeptide", "polypeptide variant", or "variant" as used herein is meant a polypeptide sequence that differs from that of a parent polypeptide sequence by virtue of at least one amino acid modification. The parent polypeptide may be a naturally occurring or wild-type (WT) polypeptide, or may be a modified version of a WT polypeptide. Variant polypeptide may refer to the polypeptide itself, a composition comprising the polypeptide, or the amino sequence that encodes it. Preferably, the variant polypeptide has at least one amino acid modification compared to the parent polypeptide, e.g. from about one to about ten amino acid modifications, and preferably from about one to about five amino acid modifications compared to the parent. The variant polypeptide sequence herein will preferably possess at least about 80% homology with a parent polypeptide sequence, and most preferably at least about 90% homology, more preferably at least about 95% homology. Accordingly, by "Fc variant" or "variant Fc" as used herein is meant an Fc sequence that differs from that of a parent Fc sequence by virtue of at least one amino acid modification. An Fc variant may only encompass an Fc region, or may exist in the context of an antibody, Fc fusion, isolated Fc, Fc fragment, or other polypeptide that is substantially encoded by Fc. Fc variant may refer to the Fc polypeptide itself, compositions comprising the Fc variant polypeptide, or the amino acid sequence that encodes it. By "Fc polypeptide variant" or "variant Fc polypeptide" as used herein is meant an Fc polypeptide that differs from a parent Fc polyeptide by virtue of at least one amino acid modification. By "Fc variant antibody" or "antibody Fc variant" as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification in the Fc region. By "protein variant" or "variant protein" as used herein is meant a protein that differs from a parent protein by virtue of at least one amino acid modification. By "antibody variant" or "variant antibody" as used herein is meant an antibody that differs from a parent antibody by virtue of at least one amino acid modification. By "IqG variant" or "variant IqG" as used herein is meant an antibody that differs from a parent IgG by virtue of at least one amino acid modification. By "immunoglobulin variant" or "variant immunoqlobluin" as used herein is meant an immunoglobulin sequence that differs from that of a parent immunoglobulin sequence by virtue of at least one amino acid modification.
[78] By "wild type" or "WT" or "native" herein is meant an amino acid sequence or a nucleotide sequence that is found in nature, including allelic variations. A WT protein, polypeptide, antibody, immunoglobulin, IgG, etc. has an amino acid sequence or a nucleotide sequence that has not been intentionally modified.
By "scFv" as used herein is meant a polypeptide consisting of two variable regions connected by a short linker sequence; e.g., VH-linker-VK, VH-linker-VA, VK-linker-VH, or VA- linker-VH. Examples of linker sequences consist of but are not limited to (Gly4Ser)3,
(Gly4Ser)4, and (Gly3Ser)4.
By "scFv-Fc" as used herein is meant an scFv fused to a Fc region; e.g., VH-linker-VK-Hinge- CH2-CH3.
[79] Immunoglobulins and Antibodies
[80] "Antibodies" (Abs) and "immunoglobulins" (Igs) are glycoproteins (although as described herein, engineered glycoforms including aglycosylated and afucosylated forms can be made) having the same structural characteristics. While antibodies exhibit binding specificity to a specific antigen, immunoglobulins include both antibodies and other antibodylike molecules which lack antigen specificity. Polypeptides of the latter kind are, for example, produced at low levels by the lymph system and at increased levels by myelomas. Thus, an immunoglobulin is a protein consisting of one or more polypeptides substantially encoded by all or part of at least one immunoglobulin gene. Immunoglobulins may have a number of structural forms, including but not limited to full length antibodies, antibody fragments and analogs (including Fc fusions and immunoadhesins as described below), as well as individual immunoglobulin domains.
[81] Antibodies are immunoglobulins that bind a specific antigen. In most mammals, including humans and mice, antibodies are constructed from paired heavy and light polypeptide chains. The light and heavy chain variable regions show significant sequence diversity between antibodies, and are responsible for binding the target antigen. Each chain is made up of individual immunoglobulin (Ig) domains, and thus the generic term
immunoglobulin can be used for such proteins.
[82] Traditional natural antibody structural units typically comprise a tetramer. Each tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one "light" (typically having a molecular weight of about 25 kDa) and one "heavy" chain (typically having a molecular weight of about 50-70 kDa). Human light chains are classified as kappa and lambda light chains. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. IgG has several subclasses, including, but not limited to lgG1 , lgG2, lgG3, and lgG4. IgM has subclasses, including, but not limited to, lgM1 and lgM2. IgA has several subclasses, including but not limited to lgA1 and lgA2. Thus, "isotype" as used herein is meant any of the classes and subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions. The known human immunoglobulin isotypes are lgG1 , lgG2, lgG3, lgG4, lgA1 , lgA2, lgM1 , lgM2, IgD, and IgE. The distinguishing features between these antibody classes are their constant regions, although subtler differences may exist in the variable region.
[83] Each of the light and heavy chains are made up of two distinct regions, referred to as the variable and constant regions. The IgG heavy chain is composed of four immunoglobulin domains linked from N- to C-terminus in the order VH-CH1 -CH2-CH3, referring to the heavy chain variable domain, heavy chain constant domain 1 , heavy chain constant domain 2, and heavy chain constant domain 3 respectively (also referred to as VH-Cy1 -Cy2-Cy3, referring to the heavy chain variable domain, constant gamma 1 domain, constant gamma 2 domain, and constant gamma 3 domain respectively). The IgG light chain is composed of two immunoglobulin domains linked from N- to C-terminus in the order VL-CL, referring to the light chain variable domain and the light chain constant domain respectively. The constant regions show less sequence diversity, and are responsible for binding a number of natural proteins to elicit important biochemical events. The structure that constitutes the natural biological form of an antibody, including the variable and constant regions, is referred to herein as a "full length antibody". In most mammals, including humans and mice, the full length antibody of the IgG isotype is a tetramer and consists of two identical pairs of two immunoglobulin chains, each pair having one light and one heavy chain, each light chain comprising immunoglobulin domains VL and CL, and each heavy chain comprising immunoglobulin domains VH, Cy1 , Cy2, and Cy3. In some mammals, for example in camels and llamas, IgG antibodies may consist of only two heavy chains, each heavy chain comprising a variable domain attached to the Fc region.
[84] The variable region of an antibody contains the antigen binding determinants of the molecule, and thus determines the specificity of an antibody for its target antigen. The variable region is so named because it is the most distinct in sequence from other antibodies within the same class. The amino-terminal portion of each chain includes a variable region of about 100 to 1 10 or more amino acids primarily responsible for antigen recognition. In the variable region, three loops are gathered for each of the V domains of the heavy chain and light chain to form an antigen-binding site. Each of the loops is referred to as a
complementarity-determining region (hereinafter referred to as a "CDR"), in which the variation in the amino acid sequence is most significant. There are 6 CDRs total, three each per heavy and light chain, designated VH CDR1 , VH CDR2, VH CDR3, VL CDR1 , VL CDR2, and VL CDR3. The variable region outside of the CDRs is referred to as the framework (FR) region. Although not as diverse as the CDRs, sequence variability does occur in the FR region between different antibodies. Overall, this characteristic architecture of antibodies provides a stable scaffold (the FR region) upon which substantial antigen binding diversity (the CDRs) can be explored by the immune system to obtain specificity for a broad array of antigens. A number of high-resolution structures are available for a variety of variable region fragments from different organisms, some unbound and some in complex with antigen.
Sequence and structural features of antibody variable regions are disclosed, for example, in Morea et al., 1997, Biophys Chem 68:9-16; Morea et al., 2000, Methods 20:267-279, entirely incorporated by reference, and the conserved features of antibodies are disclosed, for example, in Maynard et al., 2000, Annu Rev Biomed Eng 2:339-376, entirely incorporated by reference.
[85] The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function, e.g. the Fc region. In the IgG subclass of immunoglobulins, there are several immunoglobulin domains in the heavy chain referred to as heavy constant (CH) regions. In the context of IgG antibodies, the IgG isotypes each have three CH regions. Accordingly, "CH" domains in the context of IgG are as follows: "CH1 " refers to positions 1 18-220 according to the EU index as in Kabat. "CH2" refers to positions 237-340 according to the EU index as in Kabat, and "CH3" refers to positions 341 -447 according to the EU index as in Kabat.
[86] Another important region of the heavy chain is the hinge region. By "hinge" or "hinge region" or "antibody hinge region" or "immunoglobulin hinge region" herein is meant the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody. Structurally, the IgG CH1 domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237. Thus for IgG the antibody hinge is herein defined to include positions 221 (D221 in lgG1 ) to 236 (G236 in lgG1 ), wherein the numbering is according to the EU index as in Kabat. In some embodiments, for example in the context of an Fc region, the lower hinge is included, with the "lower hinge" generally referring to positions 226 or 230 to 236.
[87] For all constant region positions discussed in the present invention, numbering is according to the EU index as in Kabat (Kabat et al., 1991 , Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health, Bethesda, entirely incorporated by reference). For all variable region positions discussed in the present invention, numbering is according to the Kabat numbering scheme (Kabat et al., 1991 , ibid). Those skilled in the art of antibodies will appreciate that these conventions consist of nonsequential numbering in specific regions of an immunoglobulin sequence, enabling a normalized reference to conserved positions in immunoglobulin families. Accordingly, the positions of any given immunoglobulin as defined by the EU index or by the Kabat numbering scheme will not necessarily correspond to its sequential sequence.
[88] The present invention provides antibody analogs. By "antibody" herein is meant a protein consisting of one or more polypeptides substantially encoded by all or part of the recognized immunoglobulin genes. The recognized immunoglobulin genes, for example in humans, include the kappa (κ), lambda (λ), and heavy chain genetic loci, which together comprise the myriad variable region genes, and the constant region genes mu (υ), delta (δ), gamma (γ), sigma (σ), and alpha (a) which encode the IgM, IgD, IgG (lgG1 , lgG2, lgG3, and lgG4), IgE, and IgA (lgA1 and lgA2) isotypes respectively. Antibody herein is meant to include full length antibodies and antibody fragments, and may refer to a natural antibody from any organism, an engineered antibody, or an antibody generated recombinantly for experimental, therapeutic, or other purposes.
[89] Of interest in embodiments described herein is the antibody Fc region. The Fc region of an antibody interacts with a number of Fc receptors and ligands, imparting an array of important functional capabilities referred to as effector functions. For IgG the Fc region, Fc comprises Ig domains Cy2 and Cy3 and the N-terminal hinge leading into Cy2. An important family of Fc receptors for the IgG class are the Fc gamma receptors (FcyRs). These receptors mediate communication between antibodies and the cellular arm of the immune system (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12:181-220; Ravetch et al., 2001 , Annu Rev Immunol 19:275-290, both hereby entirely incorporated by reference). In humans this protein family includes FcyRI (CD64), including isoforms FcyRIa, FcyRIb, and FcyRIc; FcyRI I (CD32), including isoforms FcyRlla (including allotypes H 131 and R131 ), FcyRllb (including FcyRl lb- 1 and FcyRl lb-2), and FcyRl lc; and FcyRI II (CD16), including isoforms FcyRl lla (including allotypes V158 and F158) and FcyRll lb (including allotypes FcyRl l lb-NA1 and FcyRl l lb-NA2) (Jefferis et al., 2002, Immunol Lett 82:57-65, hereby entirely incorporated by reference). These receptors typically have an extracellular domain that mediates binding to Fc, a membrane spanning region, and an intracellular domain that may mediate some signaling event within the cell. These receptors are expressed in a variety of immune cells including monocytes, macrophages, neutrophils, dendritic cells, eosinophils, mast cells, platelets, B cells, large granular lymphocytes, Langerhans' cells, natural killer (NK) cells, and γδ T cells. Formation of the Fc/FcyR complex recruits these effector cells to sites of bound antigen, typically resulting in signaling events within the cells and important subsequent immune responses such as release of inflammation mediators, B cell activation,
endocytosis, phagocytosis, and cytotoxic attack. The ability to mediate cytotoxic and phagocytic effector functions is a potential mechanism by which antibodies destroy targeted cells. The cell-mediated reaction wherein nonspecific cytotoxic cells that express FcyRs recognize bound antibody on a target cell and subsequently cause lysis of the target cell is referred to as antibody dependent cell-mediated cytotoxicity (ADCC) (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12: 181 -220; Ghetie et al., 2000, Annu Rev Immunol 18:739-766; Ravetch et al., 2001 , Annu Rev Immunol 19:275-290, both hereby entirely incorporated by reference). The cell-mediated reaction wherein nonspecific cytotoxic cells that express FcyRs recognize bound antibody on a target cell and subsequently cause phagocytosis of the target cell is referred to as antibody dependent cell-mediated phagocytosis (ADCP).
[90] The different IgG subclasses have different affinities for the FcyRs, with lgG1 and lgG3 typically binding substantially better to the receptors than lgG2 and lgG4 (Jefferis et al., 2002, Immunol Lett 82:57-65, hereby entirely incorporated by reference). The FcyRs bind the IgG Fc region with different affinities. The extracellular domains of FcyRll la and FcyRl l lb are 96% identical, however FcyRll lb does not have a intracellular signaling domain.
Furthermore, whereas FcyRI, FcyRlla/c, and FcyRl lla are positive regulators of immune complex-triggered activation, characterized by having an intracellular domain that has an immunoreceptor tyrosine-based activation motif (ITAM), FcyRl lb has an immunoreceptor tyrosine-based inhibition motif (ITIM) and is therefore inhibitory. Thus the former are referred to as activation receptors, and FcyRl lb is referred to as an inhibitory receptor. Despite these differences in affinities and activities, all FcyRs bind the same region on Fc, at the N-terminal end of the Cy2 domain and the preceding hinge. [91] An overlapping but separate site on Fc serves as the interface for the complement protein C1 q. In the same way that Fc/FcyR binding mediates ADCC, Fc/C1 q binding mediates complement dependent cytotoxicity (CDC). A site on Fc between the Cy2 and Cy3 domains mediates interaction with the neonatal receptor FcRn, the binding of which recycles endocytosed antibody from the endosome back to the bloodstream (Raghavan et al., 1996, Annu Rev Cell Dev Biol 12: 181 -220; Ghetie et al., 2000, Annu Rev Immunol 18:739-766, both hereby entirely incorporated by reference). This process, coupled with preclusion of kidney filtration due to the large size of the full length molecule, results in favorable antibody serum half-lives ranging from one to three weeks. Binding of Fc to FcRn also plays a key role in antibody transport. The binding site for FcRn on Fc is also the site at which the bacterial proteins A and G bind. The tight binding by these proteins is typically exploited as a means to purify antibodies by employing protein A or protein G affinity chromatography during protein purification. The fidelity of these regions, the complement and FcRn/protein A binding regions are important for both the clinical properties of antibodies and their development.
[92] A key feature of the Fc region is the conserved N-linked glycosylation that occurs at N297. This carbohydrate, or oligosaccharide as it is sometimes referred, plays a critical structural and functional role for the antibody, and is one of the principle reasons that antibodies must be produced using mammalian expression systems. Efficient Fc binding to FcyR and C1 q requires this modification, and alterations in the composition of the N297 carbohydrate or its elimination affect binding to these proteins.
[93] The antibodies of embodiments disclosed herein may be substantially encoded by immunoglobulin genes belonging to any of the antibody classes. In certain embodiments, the antibodies disclosed herein comprise sequences belonging to the IgG class of antibodies, including lgG1 , lgG2, lgG3, or lgG4. In alternate embodiments, immunoglobulins disclosed herein comprise sequences belonging to the IgA (including subclasses lgA1 and lgA2), IgD, IgE, IgG, or IgM classes of antibodies. The immunoglobulins disclosed herein may comprise more than one protein chain, e.g., may be an antibody or Fc fusion that is a monomer or an oligomer, including a homo- or hetero-oligomer.
[94] Antibodies disclosed herein may be substantially encoded by genes from any organism, e.g., mammals (including, but not limited to humans, rodents (including but not limited to mice and rats), lagomorpha (including but not limited to rabbits and hares), camelidae (including but not limited to camels, llamas, and dromedaries), and non-human primates, including but not limited to Prosimians, Platyrrhini (New World monkeys),
Cercopithecoidea (Old World monkeys), and Hominoidea including the Gibbons and Lesser and Great Apes. In the most preferred embodiments, the immunoglobulins disclosed herein may be substantially human, with the modifications outlined herein.
[95] As is well known in the art, immunoglobulin polymorphisms exist in the human population. Gm polymorphism is determined by the IGHG1 , IGHG2 and IGHG3 genes which have alleles encoding allotypic antigenic determinants referred to as G1 m, G2m, and G3m allotypes for markers of the human lgG1 , lgG2 and lgG3 molecules (no Gm allotypes have been found on the gamma 4 chain). Markers may be classified into "allotypes" and
"isoallotypes". These are distinguished on different serological bases dependent upon the strong sequence homologies between isotypes. Allotypes are antigenic determinants specified by allelic forms of the Ig genes. Allotypes represent slight differences in the amino acid sequences of heavy or light chains of different individuals. Even a single amino acid difference can give rise to an allotypic determinant, although in many cases there are several amino acid substitutions that have occurred. Allotypes are sequence differences between alleles of a subclass whereby the antisera recognize only the allelic differences. An isoallotype is an allele in one isotype which produces an epitope which is shared with a non- polymorphic homologous region of one or more other isotypes and because of this the antisera will react with both the relevant allotypes and the relevant homologous isotypes (Clark, 1997, IgG effector mechanisms, Chem Immunol. 65:88-1 10; Gorman & Clark, 1990, Semin Immunol 2(6):457-66, both hereby entirely incorporated by reference).
[96] Allelic forms of human immunoglobulins have been well-characterized (WHO Review of the notation for the allotypic and related markers of human immunoglobulins. J
Immunogen 1976, 3: 357-362; WHO Review of the notation for the allotypic and related markers of human immunoglobulins. 1976, Eur. J. Immunol. 6, 599-601 ; Loghem E van, 1986, Allotypic markers, Monogr Allergy 19: 40-51 , all hereby entirely incorporated by reference). Additionally, other polymorphisms have been characterized (Kim et al., 2001 , J. Mol. Evol. 54:1-9, hereby entirely incorporated by reference). At present, 18 Gm allotypes are known: G1 m (1 , 2, 3, 17) or G1 m (a, x, f, z), G2m (23) or G2m (n), G3m (5, 6, 10, 1 1 , 13, 14, 15, 16, 21 , 24, 26, 27, 28) or G3m (b1 , c3, b5, bO, b3, b4, s, t, g1 , c5, u, v, g5) (Lefranc, et al., The human IgG subclasses: molecular analysis of structure, function and regulation. Pergamon, Oxford, pp. 43-78 (1990); Lefranc, G. et al., 1979, Hum. Genet.: 50, 199-21 1 , both hereby entirely incorporated by reference). Allotypes that are inherited in fixed combinations are called Gm haplotypes. The immunoglobulins disclosed herein may be substantially encoded by any allotype, isoallotype, or haplotype of any immunoglobulin gene.
[97] Nonhuman, Chimeric, Humanized, and Fully Human Antibodies [98] The variable region of an antibody, as is well known in the art, can compose sequences from a variety of species. In some embodiments, the antibody variable region can be from a nonhuman source, including but not limited to mice, rats, rabbits, camels, llamas, and monkeys. In some embodiments, the scaffold components can be a mixture from different species. As such, an antibody disclosed herein may be a chimeric antibody and/or a humanized antibody. In general, both "chimeric antibodies" and "humanized antibodies" refer to antibodies that combine regions from more than one species. For example, "chimeric antibodies" traditionally comprise variable region(s) from a mouse or other nonhuman species and the constant region(s) from a human.
[99] "Humanized antibodies" generally refer to non-human antibodies that have had the variable-domain framework regions swapped for sequences found in human antibodies. Generally in a humanized antibody the entire antibody, except the CDRs, is encoded by a polynucleotide of human origin or is identical to such an antibody except within its CDRs. The CDRs, some or all of which are encoded by nucleic acids originating in a non-human organism, are grafted into the beta-sheet framework of a human antibody variable region to create an antibody, the specificity of which is determined by the engrafted CDRs. The creation of such antibodies is described in, e.g., WO 92/1 1018, Jones, 1986, Nature
321 :522-525, Verhoeyen et al., 1988, Science 239:1534-1536. "Backmutation" of selected acceptor framework residues to the corresponding donor residues is often required to regain affinity that is lost in the initial grafted construct (U.S. Pat. No. 5,693,762, incorporated entirely by reference. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin, and thus will typically comprise a human Fc region. A variety of techniques and methods for humanizing, reshaping, and resurfacing non-human antibodies are well known in the art (See Tsurushita & Vasquez, 2004, Humanization of Monoclonal Antibodies, Molecular Biology of B Cells, 533-545, Elsevier Science (USA), and references cited therein, herein expressly incorporated by reference). In certain variations, the immunogenicity of the antibody is reduced using a method described in Lazar et al., 2007, Mol Immunol 44: 1986- 1998 and USSN 1 1 /004,590, entitled "Methods of Generating Variant Proteins with
Increased Host String Content and Compositions Thereof", filed on December 3, 2004, incorporated entirely by reference.
[100] In one embodiment, the antibody is a fully human antibody with at least one modification as outlined herein. "Fully human antibody " or "complete human antibody" refers to a human antibody having the gene sequence of an antibody derived from a human chromosome with the modifications outlined herein. Fully human antibodies may be obtained, for example, using transgenic mice (Lonberg, 2008, Handb Exp Pharmacol 181 :69-97) or human antibody libraries coupled with selection methods (Mondon et al., 2008, Front Biosci 13:1 1 17-29; Lonberg, 2008, Curr Opin Immunol. 20[4]:450-9).
[101] Antibody Analogs
[102] The present invention provides antibody analogs. As used herein, "antibodies" may comprise a variety of structures, including, but not limited to full length antibodies, antibody fragments, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as "antibody mimetics"), antibody fusions, antibody
conjugates, and fragments of each, respectively.
[103] In one embodiment, the immunogloublin comprises an antibody fragment. Specific antibody fragments include, but are not limited to (i) the Fab fragment consisting of VL, VH, CL and CH1 domains, (ii) the Fd fragment consisting of the VH and CH1 domains, (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment, which consists of a single variable, (v) isolated CDR regions, (vi) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vii) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site, (viii) bispecific single chain Fv dimers, and (ix) "diabodies" or "triabodies", multivalent or multispecific fragments
constructed by gene fusion. The antibody fragments may be modified. For example, the molecules may be stabilized by the incorporation of disulphide bridges linking the VH and VL domains. Examples of antibody formats and architectures are described in Holliger & Hudson, 2006, Nature Biotechnology 23(9):1 126-1 136, and Carter 2006, Nature Reviews Immunology 6:343-357 and references cited therein, all expressly incorporated by reference.
[104] The present invention provides antibody analogs that are a novel set of formats that enable co-engaged antigens to be bound with different valencies. Specifically, as discussed herein, there are two general formats: an antibody analog that has three antigen binding domains, and one that has two.
[105] In one preferred embodiment, a first antigen can be bound bivalently while a second antigen is simultaneously bound monovalently. The novel immunoglobulin compositions herein co-engage a first and second antigen, referred to herein as antigen-1 and antigen-2 respectively. Binding to the antigens is mediated by first and second antigen binding regions such that antigen-1 is bound bivalently by the immunoglobulin, while antigen-2 is bound monovalently by the immunoglobulin. That is, the immunoglobulin of the invention contains two binding sites for antigen-1 but only one binding site for antigen-2.
[106] In preferred embodiments of the invention, the antigen binding regions of the immunoglobulin are antibody variable regions, also referred to as Fv regions, each comprising a VH domain and a VL domain. The Fv region that binds antigen-1 is referred to as Fv-1 , while the Fv region that binds antigen-2 is referred to as Fv-2.
[107] Preferred immunoglobulin formats of the invention for co-engaging antigens with different valencies comprise IgG antibodies with a C-terminal Fv region. Such
immunoglobulins typically comprise a full length antibody that is linked C-terminally to a set of VH and VL domains that make up a novel Fv region (Fv-2) and thus a novel antigen binding site (for antigen-2). The C-terminal VH and VL domains may also be linked C- terminally to CH 1 and CL domains, thereby resulting in a C-terminal Fab that makes up the novel antigen binding site. These immunoglobulins are referred to as mAb-Fv and mAb-Fab. The VH and VL domains are typically linked C-terminally to the Fc region via flexible linkers. A variety of linkers are described herein that may find use for linking the VH and VL domains C-terminally to the constant chain in order to generate the novel binding site Fv-2.
[108] The N-terminal Fab regions of the mAb-Fv and mAb-Fab contain the binding site for the primary or first antigen (antigen-1 ). Accordingly, this Fv region is referred to as Fv-1 . Because there are two such Fv-1 regions, the mAb-Fv and mAb-Fab bind bivalently to antigen-1 . That is, one immunoglublin binds two of antigen-1 at the same time. In contrast, the C-terminal Fv region of the mAb-Fv or C-terminal Fab region of the mAb-Fab contain the binding site for the second antigen (antigen-2). Thus this Fv region is referred to as Fv-2. Because there is only one such Fv-2 region, the mAb-Fv and mAb-Fab bind monovalently to antigen-2. That is, one immunoglublin binds only one of antigen-2. The mAb-Fv and mAb- Fab immunoglobulins thus contain three variable regions, two of which are Fv-1 and one of which is Fv-2.
[109] The mAb-Fv and mAb-Fab immunoglobulins comprise three distinct immunoglobulin chains. These chains include an antibody light chain and two antibody heavy chains. One of said heavy chains comprises a VH or VH-CH 1 at its C-terminus, whereas the other of said heavy chains comprises a VL or VL-CL at its C-terminus. Thus the heavy chain dimer that is competent to bind the second antigen is a heterodimer, rather than a homodimer as is the case naturally for IgG's. The use of two distinct heavy chains is an important component of the mAb-Fv and mAb-Fab that enables the engineering of a second antigen binding site using a VH/VL pair.
[110] Because of the presence of this heterodimeric constant chain, the mAb-Fv and mAb- Fab formats are particularly suited for the use of variants that favor Fc heterodimerization and disfavor Fc homodimerization. Different variant heavy chains, for example containing CH3 substitutions, can be linked to the different heavy chains of the mAb-Fv and mAb-Fab. One variant heavy chain can be linked C-terminally to the VH of the mAb-Fv or VH-CH 1 of the mAb-Fab, while the other variant chain can be linked C-terminally to the VL of the mAb- Fv or VL-CL of the mAb-Fab, thereby generating the new monovalent Fv-2. Moreover, the additional Fv-2 VH/VL and VH-CH1 /VL-CL pairings may add further specificity to the heterodimer relative to the homodimer.
[111] Heterodimerc Fc variants are not a necessity, however, for generation and use of the mAb-Fv and mAb-Fab. Protein purification methods may be employed to purify the desired heterodimeric heavy chains. In the absence of Fc variants that favor heterodimerization and disfavor homodimerization, transfection of the three different DNAs, one light chain and two heavy chains, would result in three separate species - 25% homodimer, 50% heterodimer, and 25% homodimer. The 50% heterodimer is the species that comprises both the C- terminal VH and C-terminal VL domains, and thus the species that is comprises an Fv-2 that is competent to bind antigen-2. The two 25% homodimer species would comprise homdimeric heavy chains that contain two C-terminal VL's or two C-terminal VH's, and thus would typically not be able to bind antigen-2. Using protein purification methods described herein and generally known in the art, the desired 50% heterodimeric mAb-Fv or mAb-Fab species can be purified, yielding a composition that is competent of engaging antigen-2.
[112] The antibody analogs of the "trimeric" antigen binding domain constructs generally comprise two modified antibody heavy chains with associated light chains, where each heavy chain possesses two variable regions and a constant region. The first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain- Variable Domain-COOH, and the second modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Domain-COOH. Thus, the N- terminal variable heavy domains interact with the associated light chains to form an antigen binding site on each chain (as in a "traditional" antibody, providing two antigen binding domains). On the opposite "end" of the molecule, one of the variable domains at the C- terminus of one chain is a variable heavy domain, and the other C-terminal variable domain on the other chain is a variable light domain, such that a third antigen binding domain is formed. In most instances the two antigen binding domains at the N-terminal ends of the chains are to the same antigen, to bind the antigen bivalently. The third binding domain at the C-terminus is monovalent and generally directed to a second antigen as described herein.
[113] In some embodiments, as is depicted in Figure 8, the C-terminus of each chain may further comprise a CH1 domain as well. In this instance the first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Domain- Constant Domain (CH1 and/or CL)-COOH, and the second modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Domain-Constant Domain (CH1 and/or CL)-COOH.
[114] Also described herein are analogues of the mAb-Fv and mAb-Fab that bind both antigen-1 and antigen-2 monovalently. These immunoglobulins are referred to as Fab-Fv and Fab-Fab. Rather than comprising three separate chains, these formats contain two different heavy chains, i.e. they lack a native light chain. For one of the heavy chains, the N- terminal Fv-1 VH-CH1 half of the Fab region are replaced with the Fv-1 VL-CL region. Thus the heterodimer species of the Fab-Fv and Fab-Fab comprise an N-terminal Fab region that contains a single binding site for antigen-1 , and comprise a C-terminal Fv or Fab region that contains a single binding site for antigen-2. For the Fab-Fv the first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Heavy Domain-COOH, and the second modified antibody heavy chain has the structure NH2- Variable Light Domain-Constant Domain-Variable Light Domain. For the Fab-Fab the first modified antibody heavy chain has the structure NH2-Variable Heavy Domain-Constant Domain-Variable Heavy Domain-Constant Domain (e.g., CH1 )-COOH, and the second modified antibody heavy chain has the structure NH2-Variable Light Domain-Constant Domain-Variable Light Domain-Constant Domain (e.g., CL)-COOH.
[115] The use of VH and VL domains to generate the novel binding site Fv-2 is a preferred emodiment. However, other antigen-1 binding modalities may also be useful when linked with a C-terminal monovalent Fv-2. For example, rather than using both VH and VL domains as Fv-1 , mAb-Fv and mAb-Fab immunoglobulins may also comprise single domain VHH regions as Fv-1. Such VHH regions are described, for example, in Wesolowski et al., 2009, Med Microbiol Immunol 198[3]:157-174, and references therein, herein expressly incorporated by reference.
[116] In these constructs the N-terminal binding portion of the mAb-Fv and mAb-Fab may lack a light chain and a CH1 domain. It is also possible to utilize the C-terminal monovalent Fv-2 in the context of an Fc fusion, also referred to as an immunoadhesin. In this format, Fv- 1 is replaced with a fusion protein, for example a receptor that binds antigen-1. Thus antigen-1 is bound by a fusion protein, while antigen-2 is bound by Fv-2.
[117] Fc Modifications
[118] Antibody analogs disclosed herein may comprise an Fc variant. An Fc variant comprises one or more amino acid modifications relative to a parent Fc polypeptide, wherein the amino acid modification(s) provide one or more optimized properties. By "modification" herein is meant an alteration in the physical, chemical, or sequence properties of a protein, polypeptide, antibody, or immunoglobulin. An amino acid modification can be an amino acid substitution, insertion, and/or deletion in a polypeptide sequence. By "amino acid substitution" or "substitution" herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with another amino acid. For example, the substitution Y349T refers to a variant polypeptide, in this case a constant heavy chain variant, in which the tyrosine at position 349 is replaced with threonine. By "amino acid insertion" or "insertion" as used herein is meant the addition of an amino acid at a particular position in a parent polypeptide sequence. By "amino acid deletion" or "deletion" as used herein is meant the removal of an amino acid at a particular position in a parent polypeptide sequence.
[119] An Fc variant disclosed herein differs in amino acid sequence from its parent by virtue of at least one amino acid modification. The immunoglobulins disclosed herein may have more than one amino acid modification as compared to the parent, for example from about one to fifty amino acid modifications, e.g., from about one to ten amino acid
modifications, from about one to about five amino acid modifications, etc. compared to the parent. Thus the sequences of the Fc variants and those of the parent Fc polypeptide are substantially homologous. For example, the variant Fc variant sequences herein will possess about 80% homology with the parent Fc variant sequence, e.g., at least about 90% homology, at least about 95% homology, at least about 98% homology, at least about 99% homology, etc. Modifications disclosed herein also include glycoform modifications.
Modifications may be made genetically using molecular biology, or may be made
enzymatically or chemically.
[120] Fc variants disclosed herein are defined according to the amino acid modifications that compose them. Thus, for example, the substitution Y349T refers to a variant
polypeptide, in this case a constant heavy chain variant, in which the tyrosine at position 349 is replaced with threonine. Likewise, Y349T/T394F defines an Fc variant with the
substitutions Y349T and T394F relative to the parent Fc polypeptide. The identity of the WT amino acid may be unspecified, in which case the aforementioned variant is referred to as 349T/394F. It is noted that the order in which substitutions are provided is arbitrary, that is to say that, for example, 349T/394F is the same Fc variant as 394F/349T. Unless otherwise noted, constant region and Fc positions discussed herein are numbered according to the EU index or EU numbering scheme (Kabat et al., 1991 , Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health,
Bethesda, hereby entirely incorporated by reference). The EU index or EU index as in Kabat or EU numbering scheme refers to the numbering of the EU antibody (Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference). [121] In certain embodiments, the Fc variants disclosed herein are based on human IgG sequences, and thus human IgG sequences are used as the "base" sequences against which other sequences are compared, including but not limited to sequences from other organisms, for example rodent and primate sequences. Immunoglobulins may also comprise sequences from other immunoglobulin classes such as IgA, IgE, IgGD, IgGM, and the like. It is contemplated that, although the Fc variants disclosed herein are engineered in the context of one parent IgG, the variants may be engineered in or "transferred" to the context of another, second parent IgG. This is done by determining the "equivalent" or "corresponding" residues and substitutions between the first and second IgG, typically based on sequence or structural homology between the sequences of the first and second IgGs. In order to establish homology, the amino acid sequence of a first IgG outlined herein is directly compared to the sequence of a second IgG. After aligning the sequences, using one or more of the homology alignment programs known in the art (for example using conserved residues as between species), allowing for necessary insertions and deletions in order to maintain alignment (i.e., avoiding the elimination of conserved residues through arbitrary deletion and insertion), the residues equivalent to particular amino acids in the primary sequence of the first immunoglobulin are defined. Alignment of conserved residues may conserve 100% of such residues. However, alignment of greater than 75% or as little as 50% of conserved residues is also adequate to define equivalent residues. Equivalent residues may also be defined by determining structural homology between a first and second IgG that is at the level of tertiary structure for IgGs whose structures have been determined. In this case, equivalent residues are defined as those for which the atomic coordinates of two or more of the main chain atoms of a particular amino acid residue of the parent or precursor (N on N, CA on CA, C on C and O on O) are within about 0.13 nm, after alignment. In another embodiment, equivalent residues are within about 0.1 nm after alignment. Alignment is achieved after the best model has been oriented and positioned to give the maximum overlap of atomic coordinates of non-hydrogen protein atoms of the proteins. Regardless of how equivalent or corresponding residues are determined, and regardless of the identity of the parent IgG in which the IgGs are made, what is meant to be conveyed is that the Fc variants discovered as disclosed herein may be engineered into any second parent IgG that has significant sequence or structural homology with the Fc variant. Thus for example, if a variant antibody is generated wherein the parent antibody is human lgG1 , by using the methods described above or other methods for determining equivalent residues, the variant antibody may be engineered in another lgG1 parent antibody that binds a different antigen, a human lgG2 parent antibody, a human IgA parent antibody, a mouse lgG2a or lgG2b parent antibody, and the like. Again, as described above, the context of the parent Fc variant does not affect the ability to transfer the Fc variants disclosed herein to other parent IgGs. [122] Fc Modifications That Favor Heterodimerization
[123] Of particular benefit to the immunogloublins of the invention are Fc variants that favor heterodimerization of the Fc region relative to homodimerization. That is, by modifying one or more amino acids within each heavy chain that forms the Fc region, such that each heavy chain comprises at least one amino acid variant as compared to the other chain, binding of the different heavy chains to form heterodimeric Fc regions is favored over the individual heavy chains forming homodimers.
[124] Such variants, referred to herein as hetero-Fc variants, comprise a pair of different variants such that one pair (referred to as variant') modifies one CH3 domain (referred to as CH3'), while the other pair (variant") modifies another CH3 domain (CH3"). The variants are designed such that their interaction in a heterodimeric CH3 interface (variant'/variant") is favorable, thus resulting in a stable heterodimeric Fc species (comprising CH37CH3"). In contrast, the pairing of each variant with itself across a homodimeric CH3 interface
(variant'/variant' and variant'Vvariant") is unfavorable, resulting in unstable homodimeric Fc species (comprising CH3VCH3' and CH37CH3").
[125] For two different protein chains with identical (i.e. native) CH3 domains, in the absence of hetero-Fc variants, transfection of two different DNAs encoding two different heavy chains would result in three separate species - 25% homodimer', 50% heterodimer, and 25% homodimer". The hetero-Fc variants of the invention increase the percentage population of the heterodimer while reducing the population of the homodimers. For example, Fc variants that favor heterodimerization and disfavor homodimerization may alter the populations such that the heterodimer population increases to 70%, preferably 80%, more preferably 90%, and most preferably greater than 95%. For example, hetero-Fc variants may reduce the homodimer populations to 2.5% and 2.5% while increasing the heterodimer population to 95%. Populations of the different species may be measured using a variety of methods well-known in the art, including methods described herein, including for example gel electropheresis and biochemical assays. Heterodimeric immunogloublin species may optionally be isolated further from homodimeric species using any number of various protein purification methods well-known in the art.
[126] Hetero-Fc variants herein preferably comprise at least one substitution at a position in a CH3 domain selected from the group consisting of 349, 351 , 354, 356, 357, 364, 366, 368, 370, 392, 394, 395, 396, 397, 399, 401 , 405, 407, 409, 41 1 , and 439, wherein numbering is according to the EU index as in Kabat. In a preferred embodiment, hetero-Fc variants comprise at least one CH3 domain substitution per heavy chain selected from the group consisting of 349A, 349C, 349E, 349I, 349K, 349S, 349T, 349W, 351 E, 351 K, 354C, 356K, 357K, 364C, 364D, 364E, 364F, 364G, 364H, 364R, 364T, 364Y, 366D, 366K, 366S, 366W, 366Y, 368A, 368E, 368K, 368S, 370C, 370D, 370E, 370G, 370R, 370S, 370V, 392D, 392E, 394F, 394S, 394W, 394Y, 395T, 395V, 396T, 397E, 397S, 397T, 399K, 401 K, 405A, 405S, 407T, 407V, 409D, 409E, 41 1 D, 41 1 E, 41 1 K, and 439D. Each of these variants can be used individually or in any combination for each heavy chain Fc region. As will be appreciated by those in the art, each heavy chain can comprise different numbers of subsitutions. As shown in Tables 2 and 3, below, both heavy chains that make up the Fc region may comprise a single substitution, one chain may comprise a single substitution and the other two substitutions, both can contain two substitutions (although each chain will contain different substitutions), etc.
[127] In some embodiments, the hetero-Fc variants are made in combinations, that is, two or more variants per heavy chain Fc domain, selected from the group outlined above. Of use in particular embodiments are combination variants including, but not limited to, those shown in Tables 2 and 3 and in the Figures.
[128] Other Fc variants that favor heterodimerization that may find use in the creation of the antibody analogs of the invention have been described (Ridgeway et al., 1996, Protein Engineering 9[7]:617-621 ; US5,731 , 168; Xie et al., 2005, J Immunol Methods 296:95-101 ; Davis et al., 2010, Protein Engineering, Design & Selection 23[4]: 195-202; Gunasekaran et al., 2010, J Biol Chem 285[25]:1937-19646; PCT/US2009/000071 ; all entirely incorporated herein by reference).
[129] Fc Modifications That Modulate Binding to Fc Receptors
[130] The Fc variants disclosed herein may be optimized for improved or reduced binding to Fc receptors or Fc ligands. By "Fc receptor" or "Fc ligand" as used herein is meant a molecule, preferably a polypeptide, from any organism that binds to the Fc region of an antibody to form an Fc-ligand complex. Fc ligands include but are not limited to FcyRs, (as described above, including but not limited to FcyRllla, FcyRlla, FcyRllb, FcyRI and FcRn), C1 q, C3, mannan binding lectin, mannose receptor, staphylococcal protein A, streptococcal protein G, and viral FcyR. Fc ligands also include Fc receptor homologs (FcRH), which are a family of Fc receptors that are homologous to the FcyRs. Fc ligands may include
undiscovered molecules that bind Fc.
[131] Immunoglobulins herein may be engineered to for optimized properties, including but are not limited to enhanced or reduced affinity for an Fc receptor. By "greater affinity" or "improved affinity" or "enhanced affinity" or "better affinity" than a parent Fc polypeptide, as used herein is meant that an Fc variant binds to an Fc receptor with a significantly higher equilibrium constant of association (KA or Ka) or lower equilibrium constant of dissociation (KD or Kd) than the parent Fc polypeptide when the amounts of variant and parent polypeptide in the binding assay are essentially the same. For example, the Fc variant with improved Fc receptor binding affinity may display from about 5 fold to about 1000 fold, e.g. from about 10 fold to about 500 fold improvement in Fc receptor binding affinity compared to the parent Fc polypeptide, where Fc receptor binding affinity is determined, for example, by the binding methods disclosed herein, including but not limited to Biacore, by one skilled in the art. Accordingly, by "reduced affinity" as compared to a parent Fc polypeptide as used herein is meant that an Fc variant binds an Fc receptor with significantly lower KA or higher KD than the parent Fc polypeptide. Greater or reduced affinity can also be defined relative to an absolute level of affinity.
[132] In one embodiment, particularly useful Fc modifications for the present invention are variants that reduce or ablate binding to one or more FcyRs and/or complement proteins, thereby reducing or ablating Fc-mediated effector functions such as ADCC, ADCP, and CDC. Such variants are also referred to herein as "knockout variants" or "KO variants".
Variants that reduce binding to FcyRs and complement are useful for reducing unwanted interactions mediated by the Fc region and for tuning the selectivity of the immunoglobulins. Preferred knockout variants are described in USSN1 1/981 ,606, filed 10/31/2007, entitled "Fc Variants with Optimized Properties," herein expressly incorporated by reference. Preferred modifications include but are not limited substitutions, insertions, and deletions at positions 234, 235, 236, 237, 267, 269, 325, and 328, wherein numbering is according to the EU index. Preferred substitutions include but are not limited to 234G, 235G, 236R, 237K, 267R, 269R, 325L, and 328R, wherein numbering is according to the EU index. A preferred variant comprises 236R/328R. Variants may be used in the context of any IgG isotype or IgG isotype Fc region, including but not limited to human lgG1 , lgG2, lgG3, and/or lgG4 and combinations thereof. Preferred IgG Fc regions for reducing FcyR and complement binding and reducing Fc-mediated effector functions are lgG2 and lgG4 Fc regions. Hybrid isotypes may also be useful, for example hybrid lgG1/lgG2 isotypes as described in
USSN1 1/256,060, hereby incorporated by reference in its entirety. Other modifications for reducing FcyR and complement interactions include but are not limited to substitutions 297A, 234A, 235A, 237A, 318A, 228P, 236E, 268Q, 309L, 330S, 331 S, 220S, 226S, 229S, 238S, 233P, and 234V, as well as removal of the glycosylation at position 297 by mutational or enzymatic means or by production in organisms such as bacteria that do not glycosylate proteins. These and other modifications are reviewed in Strohl, 2009, Current Opinion in Biotechnology 20:685-691 , incorporated by reference in its entirety.
[133] Fc modifications that improve binding to FcyRs and/or complement may also find use in the immunoglobulins herein. Such Fc variants may enhance Fc-mediated effector functions such as ADCC, ADCP, and/or CDC. Preferred modifications for improving FcyR and complement binding are described in USSN1 1/124,620 (with Figure 41 being expressly incorporated by reference in its entirety for the modifications depicted therein) and
USSN1 1/396,495, expressly incorporated herein by reference. Preferred modifications comprise a substitution at a position selected from the group consisting of 236, 239, 268, 324, and 332, wherein numbering is according to the EU index. Preferred substitutions include but are not limited to 236A, 239D, 239E, 268D, 267E, 268E, 268F, 324T, 332D, and 332E. Preferred variants include but are not limited to 239D/332E, 236A/332E,
236A/239D/332E, 268F/324T, 267E/268F, 267E/324T, and 267E/268F/324T. Other modifications for enhancing FcyR and complement interactions include but are not limited to substitutions 298A, 333A, 334A, 326A, 247I, 339D, 339Q, 280H, 290S, 298D, 298V, 243L, 292P, 300L, 396L, 305I, and 396L. These and other modifications are reviewed in Strohl, 2009, ibid.
[134] In one embodiment, the immunoglobulins disclosed herein may incorporate Fc variants that enhance affinity for an inhibitory receptor FcyRllb. Such variants may provide the immunoglobulins herein with immunomodulatory activities related to FcyRllb+ cells, including for example B cells and monocytes. In one embodiment, the Fc variants provide selectively enhanced affinity to FcyRllb relative to one or more activating receptors.
Modifications for altering binding to FcyRllb are described in USSN 12/156, 183, filed May 30, 2008, entitled "Methods and Compositions for Inhibiting CD32b Expressing Cells", herein expressly incorporated by reference. In particular, Fc variants that improve binding to FcyRllb may include one or more modifications at a position selected from the group consisting of 234, 235, 236, 237, 239, 266, 267, 268, 325, 326, 327, 328, and 332, according to the EU index. Preferable substitutions for enhancing FcyRllb affinity include but are not limited to 234D, 234E, 234W, 235D, 235F, 235R, 235Y, 236D, 236N, 237D, 237N, 239D, 239E, 266M, 267D, 267E, 268D, 268E, 327D, 327E, 328F, 328W, 328Y, and 332E. More preferably, substitutions include but are not Imited to 235Y, 236D, 239D, 266M, 267E, 268D, 268E, 328F, 328W, and 328Y. Preferred Fc variants for enhancing binding to FcyRllb include but are not limited to 235Y/267E, 236D/267E, 239D/268D, 239D/267E, 267E/268D, 267E/268E, and 267E/328F.
[135] In some embodiments, immunoglobulins disclosed herein may incorporate Fc variants that improve FcRn binding. Such variants may enhance the in vivo pharmacokinetic properties of the immunoglobulins. Preferred variants that increase binding to FcRn and/or improve pharmacokinetic properties include but are not limited to substitutions at positions 259, 308, 428, and 434, including but not limited to for example 259I, 308F, 428L, 428M, 434S, 434H, 434F, 434Y, 434M, 428L/434S, 259I/308F and 259I/308F/428L (and others described in USSN12/341.769, filed Dec. 22, 2008, entitled "Fc Variants with Altered Binding to FcRn", entirely incorporated by reference). Other variants that increase Fc binding to FcRn include but are not limited to: 250E, 250Q, 428L, 428F, 250Q/428L (Hinton et al., 2004, J. Biol. Chem. 279(8): 6213-6216, Hinton et al. 2006 Journal of Immunology 176:346- 356), 256A, 272A, 286A, 305A, 307A, 307Q, 31 1A, 312A, 376A, 378Q, 380A, 382A, 434A (Shields et al, Journal of Biological Chemistry, 2001 , 276(9):6591-6604, entirely incorporated by reference), 252F, 252T, 252Y, 252W, 254T, 256S, 256R, 256Q, 256E, 256D, 256T, 309P, 31 1 S, 433R, 433S, 433I, 433P, 433Q, 434H, 434F, 434Y, 252Y/254T/256E,
433K/434F/436H, 308T/309P/31 1 S (Dall Acqua et al. Journal of Immunology, 2002, 169:5171-5180, Dall'Acqua et al., 2006, Journal of Biological Chemistry 281 :23514-23524, entirely incorporated by reference). Other modifications for modulating FcRn binding are described in Yeung et al., 2010, J Immunol, 182:7663-7671.
[136] Immunoglobulins described herein can incorporate Fc modifications in the context of any IgG isotype or IgG isotype Fc region, including but not limited to human lgG1 , lgG2, lgG3, and/or lgG4. The IgG isotype may be selected such as to alter FcyR- and/or complement- mediated effector fu notion (s). Hybrid IgG isotypes may also be useful. For example, USSN1 1/256,060 describes a number of hybrid lgG1/lgG2 constant regions that may find use in the particular invention. In some embodiments of the invention,
immunoglobulins may comprise means for isotypic modifications, that is, modifications in a parent IgG to the amino acid type in an alternate IgG. For example, an lgG1/lgG3 hybrid variant may be constructed by a substitutional means for substituting lgG1 positions in the CH2 and/or CH3 region with the amino acids from lgG3 at positions where the two isotypes differ. Thus a hybrid variant IgG antibody may be constructed that comprises one or more substitutional means, e.g., 274Q, 276K, 300F, 339T, 356E, 358M, 384S, 392N, 397M, 422I, 435R, and 436F. In other embodiments of the invention, an lgG1/lgG2 hybrid variant may be constructed by a substitutional means for substituting lgG2 positions in the CH2 and/or CH3 region with amino acids from lgG1 at positions where the two isotypes differ. Thus a hybrid variant IgG antibody may be constructed that comprises one or more substitutional means, e.g., one or more of the following amino acid substations: 233E, 234L, 235L, -236G
(referring to an insertion of a glycine at position 236), and 327A.
[137] Target Antigens
[138] The immunoglobulins of the invention may target virtually any antigen or pair of antigens. The mAb-Fv and mAb-Fab are particularly beneficial for targeting distinct antigens, that is where antigen-1 and antigen-2 are different. Thus the immunoglobulins herein preferably co-engage two target antigens. [139] Particular suitable applications of the immunoglobulins herein are co-target pairs for which it is beneficial or critical to engage a target antigen monovalently. Such antigens may be, for example, immune receptors that are activated upon immune complexation. Cellular activation of many immune receptors occurs only by cross-linking, acheived typically by antibody/antigen immune complexes, or via effector cell to target cell engagement. For some immune receptors, for example the CD3 signaling receptor on T cells, activation only upon engagement with co-engaged target is critical, as nonspecifiic cross-linking in a clinical setting can elicit a cytokine storm and toxicity. Therapeutically, by engaging such antigens monovalently rather than multivalently, using the immunoglobulins herein, such activation occurs only in response to cross-linking only in the microenvironment of the primary target antigen. The ability to target two different antigens with different valencies is a novel and useful aspect of the present invention. Examples of target antigens for which it may be therapeutically beneficial or necessary to co-engage monovalently include but are not limited to immune activating receptors such as CD3, FcyRs, toll-like receptors (TLRs) such as TLR4 and TLR9, cytokine, chemokine, cytokine receptors, and chemokine receptors.
[140] While the mAb-Fv and mAb-Fab immunoglobulins described herein are particularly beneficial for targeting distinct antigens, in some cases it may be beneficial to target only one antigen. That is, Fv-1 and Fv-2 may have specificity for the same antigen, thus binding the target antigen trivalently.
[141] Virtually any antigen may be targeted by the immunoglobulins herein, including but not limited to proteins, subunits, domains, motifs, and/or epitopes belonging to the following list of target antigens, which includes both soluble factors such as cytokines and membrane- bound factors, including transmembrane receptors: 17-IA, 4-1 BB, 4Dc, 6-keto-PGF1 a, 8-iso- PGF2a, 8-oxo-dG, A1 Adenosine Receptor, A33, ACE, ACE-2, Activin, Activin A, Activin AB, Activin B, Activin C, Activin RIA, Activin RIA ALK-2, Activin RIB ALK-4, Activin RIIA, Activin RU B, ADAM, ADAM10, ADAM 12, ADAM 15, ADAM17/TACE, ADAM8, ADAM9, ADAMTS, ADAMTS4, ADAMTS5, Addressins, aFGF, ALCAM, ALK, ALK-1 , ALK-7, alpha-1 -antitrypsin, alpha-V/beta-1 antagonist, ANG, Ang, APAF-1 , APE, APJ, APP, APRIL, AR, ARC, ART, Artemin, anti-Id, ASPARTIC, Atrial natriuretic factor, av/b3 integrin, Axl, b2M, B7-1 , B7-2, B7-H, B-lymphocyte Stimulator (BlyS), BACE, BACE-1 , Bad, BAFF, BAFF-R, Bag-1 , BAK, Bax, BCA-1 , BCAM, Bel, BCMA, BDNF, b-ECGF, bFGF, BID, Bik, BIM, BLC, BL-CAM, BLK, BMP, BMP-2 BMP-2a, BMP-3 Osteogenin, BMP-4 BMP-2b, BMP-5, BMP-6 Vgr-1 , BMP-7 (OP-1 ), BMP-8 (BMP-8a, OP-2), BMPR, BMPR-IA (ALK-3), BMPR-I B (ALK-6), BRK-2, RPK- 1 , BMPR-II (BRK-3), BMPs, b-NGF, BOK, Bombesin, Bone-derived neurotrophic factor, BPDE, BPDE-DNA, BTC, complement factor 3 (C3), C3a, C4, C5, C5a, C10, CA125, CAD- 8, Calcitonin, cAMP, carcinoembryonic antigen (CEA), carcinoma-associated antigen, Cathepsin A, Cathepsin B, Cathepsin C/DPPI, Cathepsin D, Cathepsin E, Cathepsin H, Cathepsin L, Cathepsin O, Cathepsin S, Cathepsin V, Cathepsin X/Z/P, CBL, CCI, CCK2, CCL, CCL1 , CCL1 1 , CCL12, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL2, CCL20, CCL21 , CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, CCL28, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9/10, CCR, CCR1 , CCR10, CCR10, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CD1 , CD2, CD3, CD3E, CD4, CD5, CD6, CD7, CD8, CD10, CD1 1 a, CD1 1 b, CD1 1 c, CD13, CD14, CD15, CD16, CD18, CD19, CD20, CD21 , CD22, CD23, CD25, CD27L, CD28, CD29, CD30, CD30L, CD32, CD33 (p67 proteins), CD34, CD38, CD40, CD40L, CD44, CD45, CD46, CD49a, CD52, CD54, CD55, CD56, CD61 , CD64, CD66e, CD74, CD80 (B7-1 ), CD89, CD95, CD123, CD137, CD138, CD140a, CD146, CD147, CD148, CD152, CD164, CEACAM5, CFTR, cGMP, CINC, Clostridium botulinum toxin, Clostridium perfringens toxin, CKb8-1 , CLC, CMV, CMV UL, CNTF, CNTN-1 , COX, C-Ret, CRG-2, CT-1 , CTACK, CTGF, CTLA-4, CX3CL1 , CX3CR1 , CXCL, CXCL1 , CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8, CXCL9, CXCL10, CXCL1 1 , CXCL12, CXCL13, CXCL14, CXCL15, CXCL16, CXCR, CXCR1 , CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, cytokeratin tumor-associated antigen, DAN, DCC, DcR3, DC-SIGN, Decay accelerating factor, des(1-3)-IGF-l (brain IGF-1 ), Dhh, digoxin, DNAM-1 , Dnase, Dpp, DPPIV/CD26, Dtk, ECAD, EDA, EDA-A1 , EDA-A2, EDAR, EGF, EGFR (ErbB-1 ), EMA, EMMPRIN, ENA, endothelin receptor, Enkephalinase, eNOS, Eot, eotaxinl , EpCAM, Ephrin B2/ EphB4, EPO, ERCC, E-selectin, ET-1 , Factor lla, Factor VII, Factor VI lie, Factor IX, fibroblast activation protein (FAP), Fas, FcR1 , FEN-1 , Ferritin, FGF, FGF-19, FGF-2, FGF3, FGF-8, FGFR, FGFR-3, Fibrin, FL, FLIP, Flt-3, Flt-4, Follicle stimulating hormone, Fractalkine, FZD1 , FZD2, FZD3, FZD4, FZD5, FZD6, FZD7, FZD8, FZD9, FZD10, G250, Gas 6, GCP-2, GCSF, GD2, GD3, GDF, GDF-1 , GDF-3 (Vgr-2), GDF- 5 (BMP-14, CDMP-1 ), GDF-6 (BMP-13, CDMP-2), GDF-7 (BMP-12, CDMP-3), GDF-8 (Myostatin), GDF-9, GDF-15 (MIC-1 ), GDNF, GDNF, GFAP, GFRa-1 , GFR-alpha1 , GFR- alpha2, GFR-alpha3, GITR, Glucagon, Glut 4, glycoprotein llb/llla (GP llb/llla), GM-CSF, gp130, gp72, GRO, Growth hormone releasing factor, Hapten (NP-cap or NIP-cap), HB- EGF, HCC, HCMV gB envelope glycoprotein, HCMV) gH envelope glycoprotein, HCMV UL, Hemopoietic growth factor (HGF), Hep B gp120, heparanase, Her2, Her2/neu (ErbB-2), Her3 (ErbB-3), Her4 (ErbB-4), herpes simplex virus (HSV) gB glycoprotein, HSV gD glycoprotein, HGFA, High molecular weight melanoma-associated antigen (HMW-MAA), HIV gp120, HIV NIB gp 120 V3 loop, HLA, HLA-DR, HM1 .24, HMFG PEM, HRG, Hrk, human cardiac myosin, human cytomegalovirus (HCMV), human growth hormone (HGH), HVEM, I- 309, IAP, ICAM, ICAM-1 , ICAM-3, ICE, ICOS, IFNg, Ig, IgA receptor, IgE, IGF, IGF binding proteins, IGF-1 R, IGFBP, IGF-I, IGF-II, IL, IL-1 , IL-1 R, IL-2, IL-2R, IL-4, IL-4R, IL-5, IL-5R, IL-6, IL-6R, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-18, IL-18R, IL-23, interferon (INF)-alpha, INF-beta, INF-gamma, Inhibin, iNOS, Insulin A-chain, Insulin B-chain, Insulin-like growth factor 1 , integrin alpha2, integrin alpha3, integrin alpha4, integrin alpha4/beta1 , integrin alpha4/beta7, integrin alpha5 (alphaV), integrin alpha5/beta1 , integrin alpha5/beta3, integrin alpha6, integrin betal , integrin beta2, interferon gamma, IP-10, l-TAC, JE, Kallikrein 2, Kallikrein 5, Kallikrein 6, , Kallikrein 1 1 , Kallikrein 12, Kallikrein 14, Kallikrein 15, Kallikrein L1 , Kallikrein L2, Kallikrein L3, Kallikrein L4, KC, KDR, Keratinocyte Growth Factor (KGF), laminin 5, LAMP, LAP, LAP (TGF- 1 ), Latent TGF-1 , Latent TGF-1 bp1 , LBP, LDGF, LECT2, Lefty, Lewis-Y antigen, Lewis-Y related antigen, LFA-1 , LFA-3, Lfo, LIF, LIGHT, lipoproteins, LIX, LKN, Lptn, L-Selectin, LT-a, LT-b, LTB4, LTBP-1 , Lung surfactant, Luteinizing hormone, Lymphotoxin Beta Receptor, Mac-1 , MAdCAM, MAG, MAP2, MARC, MCAM, MCAM, MCK- 2, MCP, M-CSF, MDC, Mer, METALLOPROTEASES, MGDF receptor, MGMT, MHC (HLA- DR), MIF, MIG, MIP, MIP-1 -alpha, MK, MMAC1 , MMP, MMP-1 , MMP-10, MMP-1 1 , MMP-12, MMP-13, MMP-14, MMP-15, MMP-2, MMP-24, MMP-3, MMP-7, MMP-8, MMP-9, MPIF, Mpo, MSK, MSP, mucin (Mud ), MUC18, Muellerian-inhibitin substance, Mug, MuSK, NAIP, NAP, NCAD, N-Cadherin, NCA 90, NCAM, NCAM, Neprilysin, Neurotrophin-3,-4, or -6, Neurturin, Neuronal growth factor (NGF), NGFR, NGF-beta, nNOS, NO, NOS, Npn, NRG-3, NT, NTN, OB, OGG1 , OPG, OPN, OSM, OX40L, OX40R, p150, p95, PADPr, Parathyroid hormone, PARC, PARP, PBR, PBSF, PCAD, P-Cadherin, PCNA, PDGF, PDGF, PDK-1 , PECAM, PEM, PF4, PGE, PGF, PGI2, PGJ2, PIN, PLA2, placental alkaline phosphatase (PLAP), PIGF, PLP, PP14, Proinsulin, Prorelaxin, Protein C, PS, PSA, PSCA, prostate specific membrane antigen (PSMA), PTEN, PTHrp, Ptk, PTN, R51 , RANK, RANKL,
RANTES, RANTES, Relaxin A-chain, Relaxin B-chain, renin, respiratory syncytial virus (RSV) F, RSV Fgp, Ret, Rheumatoid factors, RLIP76, RPA2, RSK, S100, SCF/KL, SDF-1 , SERINE, Serum albumin, sFRP-3, Shh, SIGIRR, SK-1 , SLAM, SLPI, SMAC, SMDF, SMOH, SOD, SPARC, Stat, STEAP, STEAP-II, TACE, TACI, TAG-72 (tumor-associated
glycoprotein-72), TARC, TCA-3, T-cell receptors (e.g., T-cell receptor alpha/beta), TdT, TECK, TEM1 , TEM5, TEM7, TEM8, TERT, testicular PLAP-like alkaline phosphatase, TfR, TGF, TGF-alpha, TGF-beta, TGF-beta Pan Specific, TGF-beta Rl (ALK-5), TGF-beta Rll, TGF-beta Rllb, TGF-beta Rill, TGF-beta1 , TGF-beta2, TGF-beta3, TGF-beta4, TGF-beta5, Thrombin, Thymus Ck-1 , Thyroid stimulating hormone, Tie, TIMP, TIQ, Tissue Factor, TMEFF2, Tmpo, TMPRSS2, TNF, TNF-alpha, TNF-alpha beta, TNF-beta2, TNFc, TNF-RI, TNF-RII, TNFRSF10A (TRAIL R1 Apo-2, DR4), TNFRSF10B (TRAIL R2 DR5, KILLER, TRICK-2A, TRICK-B), TNFRSF10C (TRAIL R3 DcR1 , LIT, TRID), TNFRSF10D (TRAIL R4 DcR2, TRUNDD), TNFRSF1 1 A (RANK ODF R, TRANCE R), TNFRSF1 1 B (OPG OCIF, TR1 ), TNFRSF12 (TWEAK R FN14), TNFRSF13B (TACI), TNFRSF13C (BAFF R),
TNFRSF14 (HVEM ATAR, HveA, LIGHT R, TR2), TNFRSF16 (NGFR p75NTR), TNFRSF17 (BCMA), TNFRSF18 (GITR AITR), TNFRSF19 (TROY TAJ, TRADE), TNFRSF19L (RELT), TNFRSF1A (TNF Rl CD120a, p55-60), TNFRSF1 B (TNF Rl l CD 120b, p75-80), TNFRSF26 (TNFRH3), TNFRSF3 (LTbR TNF Ri ll, TNFC R), TNFRSF4 (OX40 ACT35, TXGP1 R), TNFRSF5 (CD40 p50), TNFRSF6 (Fas Apo-1 , APT1 , CD95), TN FRSF6B (DcR3 M68, TR6), TNFRSF7 (CD27), TNFRSF8 (CD30), TNFRSF9 (4-1 BB CD137, I LA), TNFRSF21 (DR6), TNFRSF22 (DcTRAI L R2 TNFRH2), TNFRST23 (DcTRAI L R1 TNFRH 1 ), TNFRSF25 (DR3 Apo-3, LARD, TR-3, TRAMP, WSL-1 ), TN FSF10 (TRAIL Apo-2 Ligand, TL2), TNFSF1 1 (TRANCE/RANK Ligand ODF, OPG Ligand), TNFSF12 (TWEAK Apo-3 Ligand, DR3 Ligand), TNFSF13 (APRI L TALL2), TNFSF13B (BAFF BLYS, TALL1 , THANK, TNFSF20), TNFSF14 (LIGHT HVEM Ligand, LTg), TNFSF15 (TL1A/VEGI), TNFSF18 (GITR Ligand AITR Ligand, TL6), TNFSF1 A (TN F-a Conectin, DI F, TNFSF2), TN FSF1 B (TNF-b LTa, TNFSF1 ), TNFSF3 (LTb TNFC, p33), TN FSF4 (OX40 Ligand gp34, TXGP1 ), TNFSF5 (CD40 Ligand CD 154, gp39, HIGM1 , IMD3, TRAP), TNFSF6 (Fas Ligand Apo-1 Ligand, APT1 Ligand), TNFSF7 (CD27 Ligand CD70), TNFSF8 (CD30 Ligand CD153), TNFSF9 (4- 1 BB Ligand CD137 Ligand), TP-1 , t-PA, Tpo, TRAIL, TRAI L R, TRAI L-R1 , TRAI L-R2, TRANCE, transferring receptor, TRF, Trk, TROP-2, TSG, TSLP, tumor-associated antigen CA 125, tumor-associated antigen expressing Lewis Y related carbohydrate, TWEAK, TXB2, Ung, uPAR, uPAR-1 , Urokinase, VCAM, VCAM-1 , VECAD, VE-Cadherin, VE-cadherin-2, VEFGR-1 (flt-1 ), VEGF, VEGFR, VEGFR-3 (flt-4), VEGI, VIM, Viral antigens, VLA, VLA-1 , VLA-4, VN R integrin, von Willebrands factor, WI F-1 , WNT1 , WNT2, WNT2B/13, WNT3, WNT3A, WNT4, WNT5A, WNT5B, WNT6, WNT7A, WNT7B, WNT8A, WNT8B, WNT9A, WNT9A, WNT9B, WNT10A, WNT10B, WNT1 1 , WNT16, XCL1 , XCL2, XCR1 , XCR1 , XEDAR, XIAP, XPD, and receptors for hormones and growth factors.
[142] Exemplary antigens that may be targeted specifically by the immunoglobulins of the invention include but are not limited to: CD20, CD19, Her2, EGFR, EpCAM, CD3, FcyRl lla (CD16), FcyRl la (CD32a), FcyRl lb (CD32b), FcyRI (CD64), Toll-like receptors (TLRs) such as TLR4 and TLR9, cytokines such as I L-2, IL-5, I L-13, I L-12, IL-23, and TNFa, cytokine receptors such as I L-2R, chemokines, chemokine receptors, growth factors such as VEGF and HGF, and the like.
[143] The choice of suitable target antigens and co-targets depends on the desired therapeutic application. Some targets that have proven especially amenable to antibody therapy are those with signaling functions. Other therapeutic antibodies exert their effects by blocking signaling of the receptor by inhibiting the binding between a receptor and its cognate ligand. Another mechanism of action of therapeutic antibodies is to cause receptor down regulation. Other antibodies do not work by signaling through their target antigen. The choice of co-targets will depend on the detailed biology underlying the pathology of the indication that is being treated. [144] Monoclonal antibody therapy has emerged as an important therapeutic modality for cancer (Weiner et al., 2010, Nature Reviews Immunology 10:317-327; Reichert et al., 2005, Nature Biotechnology 23[9]: 1073-1078; herein expressly incorporated by reference). For anti-cancer treatment it may be desirable to target one antigen (antigen-1 ) whose expression is restricted to the cancerous cells while co-targeting a second antigen (antigen-2) that mediates some immunulogical killing activity. For other treatments it may be beneficial to co- target two antigens, for example two angiogenic factors or two growth factors that are each known to play some role in proliferation of the tumor. Exemplary co-targets for oncology include but are not limited to HGF and VEGF, IGF-1 R and VEGF, Her2 and VEGF, CD19 and CD3, CD20 and CD3, Her2 and CD3, CD19 and FcyRN Ia, CD20 and FcyRN Ia, Her2 and FcyRNIa. An immunoglobulin of the invention may be capable of binding VEGF and phosphatidylserine; VEGF and ErbB3; VEGF and PLGF; VEGF and ROB04; VEGF and BSG2; VEGF and CDCP1 ; VEGF and ANPEP; VEGF and c-MET; HER-2 and ERB3; HER-2 and BSG2; HER-2 and CDCP1 ; H ER-2 and ANPEP; EGFR and CD64; EGFR and BSG2; EGFR and CDCP1 ; EGFR and ANPEP; IGF1 R and PDGFR; IGF1 R and VEGF; IGF1 R and CD20; CD20 and CD74; CD20 and CD30; CD20 and DR4; CD20 and VEGFR2; CD20 and CD52; CD20 and CD4; HGF and c-MET; HGF and NRP1 ; HGF and phosphatidylserine; ErbB3 and IGF1 R; ErbB3 and IGF1 ,2; c-Met and Her-2; c-Met and NRP1 ; c-Met and IGF1 R; IGF1 ,2 and PDGFR; IGF1 ,2 and CD20; IGF1 ,2 and IGF1 R; IGF2 and EGFR; IGF2 and HER2; IGF2 and CD20; IGF2 and VEGF; IGF2 and IGF1 R; IGF1 and IGF2; PDGFRa and VEGFR2; PDGFRa and PLGF; PDGFRa and VEGF; PDGFRa and c-Met; PDGFRa and EGFR; PDGFRb and VEGFR2; PDGFRb and c-Met; PDGFRb and EGFR; RON and c-Met; RON and MTSP1 ; RON and MSP; RON and CDCP1 ; VGFR1 and PLGF; VGFR1 and RON; VGFR1 and EGFR; VEGFR2 and PLGF; VEGFR2 and NRP1 ; VEGFR2 and RON; VEGFR2 and DLL4; VEGFR2 and EGFR; VEGFR2 and ROB04; VEGFR2 and CD55; LPA and S1 P; EPHB2 and RON; CTLA4 and VEGF; CD3 and EPCAM; CD40 and IL6; CD40 and IGF; CD40 and CD56; CD40 and CD70; CD40 and VEGFR1 ; CD40 and DR5; CD40 and DR4; CD40 and APRI L; CD40 and BCMA; CD40 and RANKL; CD28 and MAPG; CD80 and CD40; CD80 and CD30; CD80 and CD33; CD80 and CD74; CD80 and CD2; CD80 and CD3; CD80 and CD19; CD80 and CD4; CD80 and CD52; CD80 and VEGF; CD80 and DR5; CD80 and VEGFR2; CD22 and CD20; CD22 and CD80; CD22 and CD40; CD22 and CD23; CD22 and CD33; CD22 and CD74; CD22 and CD19; CD22 and DR5; CD22 and DR4; CD22 and VEGF; CD22 and CD52; CD30 and CD20; CD30 and CD22; CD30 and CD23; CD30 and CD40; CD30 and VEGF; CD30 and CD74; CD30 and CD19; CD30 and DR5; CD30 and DR4; CD30 and VEGFR2; CD30 and CD52; CD30 and CD4; CD138 and RANKL; CD33 and FTL3; CD33 and VEGF; CD33 and VEGFR2; CD33 and CD44; CD33 and DR4; CD33 and DR5; DR4 and CD137; DR4 and IGF1.2; DR4 and IGF1R; DR4 and DR5; DR5 and CD40; DR5 and CD137; DR5 and CD20; DR5 and EGFR; DR5 and IGF1.2; DR5 and IGFR, DR5 and HER-2, and EGFR and DLL4. Other target combinations include one or more members of the EGF/erb-2/erb-3 family.
[145] Other targets (one or more) involved in oncological diseases that the
immunoglobulins herein may bind include, but are not limited to those selected from the group consisting of: CD52, CD20, CD19, CD3, CD4, CD8, BMP6, IL12A, IL1A, IL1B, 1L2, IL24, INHA, TNF, TNFSF10, BMP6, EGF, FGF1, FGF10, FGF11, FGF12, FGF13, FGF14, FGF16, FGF17, FGF18, FGF19, FGF2, FGF20, FGF21, FGF22, FGF23, FGF3, FGF4, FGF5, FGF6, FGF7, FGF8, FGF9, GRP, IGF1, IGF2, IL12A, IL1A, IL1B, 1L2, INHA, TGFA, TGFB1, TGFB2, TGFB3, VEGF, CDK2, FGF10, FGF18, FGF2, FGF4, FGF7, IGF1R, IL2, BCL2, CD164, CDKN1A, CDKN1B, CDKN1C, CDKN2A, CDKN2B, CDKN2C, CDKN3, GNRH1, IGFBP6, IL1A, IL1B, ODZ1, PAWR, PLG, TGFB1I1, AR, BRCA1, CDK3, CDK4, CDK5, CDK6, CDK7, CDK9, E2F1, EGFR, EN01, ERBB2, ESR1, ESR2, IGFBP3, IGFBP6, IL2, INSL4, MYC, NOX5, NR6A1, PAP, PCNA, PRKCQ, PRKD1, PRL, TP53, FGF22, FGF23, FGF9, IGFBP3, IL2, INHA, KLK6, TP53, CHGB, GNRH1, IGF1, IGF2, INHA, INSL3, INSL4, PRL, KLK6, SHBG, NR1D1, NR1H3, NR1I3, NR2F6, NR4A3, ESR1, ESR2, NR0B1, NR0B2, NR1D2, NR1H2, NR1H4, NR112, NR2C1, NR2C2, NR2E1, NR2E3, NR2F1, NR2F2, NR3C1, NR3C2, NR4A1, NR4A2, NR5A1, NR5A2, NR6 μΙ, PGR, RARB, FGF1, FGF2, FGF6, KLK3, KRT1, APOC1, BRCA1, CHGA, CHGB, CLU, COL1A1, COL6A1, EGF, ERBB2, ERK8, FGF1, FGF10, FGF11, FGF13, FGF14, FGF16, FGF17, FGF18, FGF2, FGF20, FGF21, FGF22, FGF23, FGF3, FGF4, FGF5, FGF6, FGF7, FGF8, FGF9, GNRH1, IGF1, IGF2, IGFBP3, IGFBP6, IL12A, IL1A, IL1B, 1L2, IL24, INHA, INSL3, INSL4, KLK10, KLK12, KLK13, KLK14, KLK15, KLK3, KLK4, KLK5, KLK6, KLK9, MMP2, MMP9, MSMB, NTN4, ODZ1, PAP, PLAU, PRL, PSAP, SERPINA3, SHBG, TGFA, TIMP3, CD44, CDH1, CDH10, CDH19, CDH20, CDH7, CDH9, CDH1, CDH10, CDH13, CDH18, CDH19, CDH20, CDH7, CDH8, CDH9, ROB02, CD44, ILK, ITGA1, APC, CD164, COL6A1, MTSS1, PAP, TGFB1I1, AGR2, AIG1, AKAP1, AKAP2, CANT1, CAV1, CDH12, CLDN3, CLN3, CYB5, CYC1, DAB21P, DES, DNCL1, ELAC2, EN02, EN03, FASN, FLJ12584, FLJ25530, GAGEB1, GAGEC1, GGT1, GSTP1, HIP1, HUMCYT2A, IL29, K6HF, KAI1, KRT2A, MIB1, PART1, PATE, PCA3, PIAS2, PIK3CG, PPID, PR1, PSCA, SLC2A2, SLC33 μΙ, SLC43 μΙ, STEAP, STEAP2, TPM1, TPM2, TRPC6, ANGPT1, ANGPT2, ANPEP, ECGF1, EREG, FGF1, FGF2, FIGF, FLT1, JAG1, KDR, LAMA5, NRP1, NRP2, PGF, PLXDC1, STAB 1, VEGF, VEGFC, ANGPTL3, BAM, COL4A3, IL8, LAMA5, NRP1, NRP2, STAB 1, ANGPTL4, PECAM1, PF4, PROK2, SERPINF1, TNFAIP2, CCL11, CCL2, CXCL1, CXCL10, CXCL3, CXCL5, CXCL6, CXCL9, IFNA1, IFNB1, IFNG, IL1B, IL6, MDK, EDG1, EFNA1, EFNA3, EFNB2, EGF, EPHB4, FGFR3, HGF, IGF1 , ITGB3, PDGFA, TEK, TGFA, TGFB1 , TGFB2, TGFBR1 , CCL2, CDH5, C0L1A1 , EDG1 , ENG, ITGAV, ITGB3, THBS1 , THBS2, BAD, BAG1 , BCL2, CCNA1 , CCNA2, CCND1 , CCNE1 , CCNE2, CDH1 (E-cadherin), CDKN1 B (p27Kip1 ), CDKN2A (p161 NK4a), C0L6A1 , CTNNB1 (b-catenin), CTSB (cathepsin B), ERBB2 (Her-2), ESR1 , ESR2, F3 (TF), F0SL1 (FRA-1 ), GAT A3, GSN (Gelsolin), IGFBP2, IL2RA, IL6, IL6R, IL6ST (glycoprotein 130), ITGA6 (a6 integrin), JUN, KLK5, KRT19, MAP2K7 (c-Jun), MKI67 (Ki-67), NGFB (GF), NGFR, NME1 (M23A), PGR, PLAU (uPA), PTEN, SERPINB5 (maspin), SERPINE1 (PAI-1 ), TGFA, THBS1 (thrombospondin-1 ), TIE (Tie-1 ), TNFRSF6 (Fas), TNFSF6 (FasL), TOP2A (topoisomerase lia), TP53, AZGP1 (zinc- a-glycoprotein), BPAG1 (plectin), CDKN1A (p21 Wap1/Cip1 ), CLDN7 (claudin-7), CLU (clusterin), ERBB2 (Her-2), FGF1 , FLRT1 (fibronectin), GABRP (GABAa), GNAS1 , ID2, ITGA6 (a6 integrin), ITGB4 (b 4 integrin), KLF5 (GC Box BP), KRT19 (Keratin 19), KRTHB6 (hair-specific type II keratin), MACMARCKS, MT3 (metallothionectin-lll), MUC1 (mucin), PTGS2 (COX-2), RAC2 (p21 Rac2), S100A2, SCGB1 D2 (lipophilin B), SCGB2A1
(mammaglobin 2), SCGB2A2 (mammaglobin 1 ), SPRR1 B (Spr1 ), THBS1 , THBS2, THBS4, and TNFAIP2 (B94), RON, c-Met, CD64, DLL4, PLGF, CTLA4, phophatidylserine, ROB04, CD80, CD22, CD40, CD23, CD28, CD80, CD55, CD38, CD70, CD74, CD30, CD138, CD56, CD33, CD2, CD137, DR4, DR5, RANKL, VEGFR2, PDGFR, VEGFR1 , MTSP1 , MSP, EPHB2, EPHA1 , EPHA2, EpCAM, PGE2, NKG2D, LPA, SIP, APRIL, BCMA, MAPG, FLT3, PDGFR alpha, PDGFR beta, ROR1 , PSMA, PSCA, SCD1 , and CD59.
[146] Monoclonal antibody therapy has become an important therapeutic modality for treating autoimmune and inflammatory disorders (Chan & Carter, 2010, Nature Reviews Immunology 10:301 -316; Reichert et al., 2005, Nature Biotechnology 23[9]:1073-1078; herein expressly incorporated by reference). Many proteins have been implicated in general autoimmune and inflammatory responses, and thus may be targeted by the immunogloublins of the invention. Autoimmune and inflammatory targets include but are not limited to C5, CCL1 (I-309), CCL1 1 (eotaxin), CCL13 (mcp-4), CCL15 (MIP-1 d), CCL16 (HCC-4), CCL17 (TARC), CCL18 (PARC), CCL19, CCL2 (mcp-1 ), CCL20 (MIP-3a), CCL21 (MIP-2), CCL23 (MPIF-1 ), CCL24 (MPIF-2/eotaxin-2), CCL25 (TECK), CCL26, CCL3 (MIP-1 a), CCL4 (MIP- 1 b), CCL5 (RANTES), CCL7 (mcp-3), CCL8 (mcp-2), CXCL1 , CXCL10 (IP-10), CXCL1 1 (1 - TAC/IP-9), CXCL12 (SDF1 ), CXCL13, CXCL14, CXCL2, CXCL3, CXCL5 (ENA-78/LIX), CXCL6 (GCP-2), CXCL9, IL13, IL8, CCL13 (mcp-4), CCR1 , CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CX3CR1 , IL8RA, XCR1 (CCXCR1 ), IFNA2, IL10, IL13, IL17C, IL1A, IL1 B, IL1 F10, IL1 F5, IL1 F6, IL1 F7, IL1 F8, IL1 F9, IL22, IL5, IL8, IL9, LTA, LTB, MIF, SCYE1 (endothelial Monocyte-activating cytokine), SPP1 , TNF, TNFSF5, IFNA2, IL10RA, IL10RB, IL13, IL13RA1 , IL5RA, IL9, IL9R, ABCF1 , BCL6, C3, C4A, CEBPB, CRP, ICEBERG, IL1R1, IL1RN, IL8RB, LTB4R, TOLLIP, FADD, IRAKI, IRAK2, MYD88, NCK2, TNFAIP3, TRADD, TRAF1, TRAF2, TRAF3, TRAF4, TRAF5, TRAF6, ACVR1, ACVR1B, ACVR2, ACVR2B, ACVRL1, CD28, CD3E, CD3G, CD3Z, CD69, CD80, CD86, CNR1, CTLA4, CYSLTR1, FCER1A, FCER2, FCGR3A, GPR44, HAVCR2, OPRD1, P2RX7, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, BLR1, CCL1, CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CX3CL1, CX3CR1, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL10, CXCL11, CXCL12, CXCL13, CXCR4, GPR2, SCYE1, SDF2, XCL1, XCL2, XCR1, AMH, AMHR2, BMPR1A, BMPR1B, BMPR2, C19orf10 (IL27w), CER1, CSF1, CSF2, CSF3, DKFZp451J0118, FGF2, GFI1, IFNA1, IFNB1, IFNG, IGF1, IL1A, IL1B, IL1R1, IL1R2, IL2, IL2RA, IL2RB, IL2RG, IL3, IL4, IL4R, IL5, IL5RA, IL6, IL6R, IL6ST, IL7, IL8, IL8RA, IL8RB, IL9, IL9R, IL10, IL10RA, IL10RB, IL11, IL12RA, IL12A, IL12B, IL12RB1, IL12RB2, IL13, IL13RA1, IL13RA2, IL15, IL15RA, IL16, IL17, IL17R, IL18, IL18R1, IL19, IL20, KITLG, LEP, LTA, LTB, LTB4R, LTB4R2, LTBR, MIF, NPPB, PDGFB, TBX21, TDGF1, TGFA, TGFB1, TGFB1I1, TGFB2, TGFB3, TGFB1, TGFBR1 , TGFBR2, TGFBR3, TH1L, TNF, TNFRSF1A, TNFRSF1B, TNFRSF7, TNFRSF8, TNFRSF9, TNFRSF11A, TNFRSF21, TNFSF4,
TNFSF5, TNFSF6, TNFSF11, VEGF, ZFPM2, and RNF110 (ZNF144).
[147] Exemplary co-targets for autoimmune and inflammatory disorders include but are not limited to IL-1 and TNFalpha, IL-6 and TNFalpha, IL-6 and IL-1, IgE and IL-13, IL-1 and IL- 13, IL-4 and IL-13, IL-5 and IL-13, IL-9 and IL-13, CD19 and FcyRllb, and CD79 and
FcyRllb.
[148] Immunglobulins of the invention with specificity for the following pairs of targets to treat inflammatory disease are contemplated: TNF and IL-17A; TNF and RANKL; TNF and VEGF; TNF and SOST; TNF and DKK; TNF and alphaVbeta3; TNF and NGF; TNF and IL- 23p19; TNF and IL-6; TNF and SOST; TNF and IL-6R; TNF and CD-20; IgE and IL-13; IL-13 and IL23p19; IgE and IL-4; IgE and IL-9; IgE and IL-9; IgE and IL-13; IL-13 and IL-9; IL-13 and IL-4; IL-13 and IL-9; IL-13 and IL-9; IL-13 and IL-4; IL-13 and IL-23p19; IL-13 and IL-9; IL-6R and VEGF; IL-6Rand IL-17A; IL-6Rand RANKL; IL-17A and IL-1beta; IL-1betaand RANKL; IL-1beta and VEGF; RANKL and CD-20; IL-1alpha and IL-1beta; IL-1alpha and IL- 1beta.
[149] Pairs of targets that the immunoglobulins described herein can bind and be useful to treat asthma may be determined. In an embodiment, such targets include, but are not limited to, IL-13 and IL-1 beta, since IL-1 beta is also implicated in inflammatory response in asthma; IL-13 and cytokines and chemokines that are involved in inflammation, such as IL-13 and IL- 9; IL-13 and IL-4; IL-13 and IL-5; IL-13 and IL-25; IL-13 and TARC; IL-13 and MDC; IL-13 and MIF; IL-13 and TGF-β; IL-13 and LHR agonist; IL-13 and CL25; IL-13 and SPRR2a; IL- 13 and SPRR2b; and IL-13 and ADAM8. The immunoglobulins herein may have specifity for one or more targets involved in asthma selected from the group consisting of CSF1 (MCSF), CSF2 (GM-CSF), CSF3 (GCSF), FGF2, IFNA1 , IFNB1 , IFNG, histamine and histamine receptors, IL1A, IL1 B, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL1 1 , IL12A, IL12B, IL13, IL14, IL15, IL16, IL17, IL18, IL19, KITLG, PDGFB, IL2RA, IL4R, IL5RA, IL8RA, IL8RB, IL12RB1 , IL12RB2, IL13RA1 , IL13RA2, IL18R1 , TSLP, CCLi, CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL13, CCL17, CCL18, CCL19, CCL20, CCL22, CCL24,CX3CL1 , CXCL1 , CXCL2, CXCL3, XCLi, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CX3CR1 , GPR2, XCR1 , FOS, GAT A3, JAK1 , JAK3, STAT6, TBX21 , TGFB1 , TNF, TNFSF6, YY1 , CYSLTR1 , FCER1A, FCER2, LTB4R, TB4R2, LTBR, and Chitinase.
[150] Pairs of targets involved in rheumatoid arthritis (RA) may be co-targeted by the invention, including but not limited to TNF and IL-18; TNF and IL-12; TNF and IL-23; TNF and IL-1 beta; TNF and MIF; TNF and IL-17; and TNF and IL-15.
[151] Antigens that may be targeted in order to treat systemic lupus erythematosus (SLE) by the immunoglobulins herein include but are not limited to CD-20, CD-22, CD-19, CD28, CD4, CD80, HLA-DRA, IL10, IL2, IL4, TNFRSF5, TNFRSF6, TNFSF5, TNFSF6, BLR1 , HDAC4, HDAC5, HDAC7A, HDAC9, ICOSL, IGBP1 , MS4A1 , RGSI, SLA2, CD81 , IFNB1 , IL10, TNFRSF5, TNFRSF7, TNFSF5, AICDA, BLNK, GALNAC4S-6ST, HDAC4, HDAC5, HDAC7A, HDAC9, IL10, IL1 1 , IL4, INHA, INHBA, KLF6, TNFRSF7, CD28, CD38, CD69, CD80, CD83, CD86, DPP4, FCER2, IL2RA, TNFRSF8, TNFSF7, CD24, CD37, CD40, CD72, CD74, CD79A, CD79B, CR2, ILIR2, ITGA2, ITGA3, MS4A1 , ST6GALI, CDIC, CHSTIO, HLA-A, HLA-DRA, and NT5E.; CTLA4, B7.1 , B7.2, BlyS, BAFF, C5, IL-4, IL-6, IL- 10, IFN-a, and TNF-a.
[152] The immunoglobulins herein may target antigens for the treatment of multiple sclerosis (MS), inlcuding but not limited to IL-12, TWEAK, IL-23, CXCL13, CD40, CD40L, IL- 18, VEGF, VLA-4, TNF, CD45RB, CD200, IFNgamma, GM-CSF, FGF, C5, CD52, and CCR2. An embodiment includes co-egagement of anti-IL-12 and TWEAK for the treatment of MS.
[153] One aspect of the invention pertains to immunoglobulins capable of binding one or more targets involved in sepsis, in an embodiment two targets, selected from the group consisting TNF, IL-1 , MIF, IL-6, IL-8, IL-18, IL-12, IL-23, FasL, LPS, Toll-like receptors, TLR- 4, tissue factor, MIP-2, ADORA2A, CASP1 , CASP4, IL-10, IL-1 B, N FKB1 , PROC,
TN FRSFIA, CSF3, CCR3, I LI RN, MI F, N FKB1 , PTAFR, TLR2, TLR4, GPR44, HMOX1 , midkine, I RAKI , N FKB2, SERPI NA1 , SERPI N E1 , and TREM 1 . [154] In some cases, immunoglobulins herein may be directed against antigens for the treatment of infectious diseases.
[155] One skilled in the art will appreciate that any of the aforementioned target antigens, the ligands or receptors that bind them, or other members of their corresponding biochemical pathway, may be operably linked to the immunoglobulins of the present invention in order to generate an Fc fusion. Thus for example, an Fc fusion that targets TNFa could be constructed by operably linking an Fc region to TNFR1 or TNFR2. Thus virtually any polypeptide, whether a ligand, receptor, or some other protein or protein domain, including but not limited to the aforementioned targets and the proteins that compose their
corresponding biochemical pathways, may be operably linked to the immunoglobulins of the present invention to develop an Fc fusion.
[156] Linkers
[157] The term "linker" is used to denote polypeptides comprising two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions. Such linker polypeptides are well known in the art (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1 121-1 123). A variety of linkers may find use in some embodiments described herein to covalently link Fc regions to a fusion partner. "Linker" herein is also referred to as "linker sequence", "spacer", "tethering sequence" or grammatical equivalents thereof. Homo-or hetero-bifunctional linkers as are well known (see, 1994 Pierce Chemical Company catalog, technical section on cross- linkers, pages 155-200, incorporated entirely by reference). A number of strategies may be used to covalently link molecules together. These include, but are not limited to polypeptide linkages between N- and C-termini of proteins or protein domains, linkage via disulfide bonds, and linkage via chemical cross-linking reagents. In one aspect of this embodiment, the linker is a peptide bond, generated by recombinant techniques or peptide synthesis. The linker peptide may predominantly include the following amino acid residues: Gly, Ser, Ala, or Thr. The linker peptide should have a length that is adequate to link two molecules in such a way that they assume the correct conformation relative to one another so that they retain the desired activity. In one embodiment, the linker is from about 1 to 50 amino acids in length, preferably about 1 to 30 amino acids in length. In one embodiment, linkers of 1 to 20 amino acids in length may be used. Useful linkers include glycine-serine polymers, including for example (GS)n, (GSGGS)n, (GGGGS)n, and (GGGS)n, where n is an integer of at least one, glycine-alanine polymers, alanine-serine polymers, and other flexible linkers.
Alternatively, a variety of nonproteinaceous polymers, including but not limited to
polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol, may find use as linkers, that is may find use as linkers.
[158] Other linker sequences may include any sequence of any length of CL/CH 1 domain but not all residues of CL/CH 1 domain; for example the first 5-12 amino acid residues of the CL/CH 1 domains. Linkers can be derived from immunoglobulin light chain, for example CK or C . Linkers can be derived from immunoglobulin heavy chains of any isotype, including for example Cy1 , Cy2, Cy3, Cy4, Ca1 , Ca2, C8, Cs, and Ομ. Linker sequences may also be derived from other proteins such as Ig-like proteins (e.g. TCR, FcR, KI R), hinge region- derived sequences, and other natural sequences from other proteins.
[159] Glvcoform Modifications
[160] All antibodies contain carbohydrate at conserved positions in the constant regions of the heavy chain. Each antibody isotype has a distinct variety of N-linked carbohydrate structures. Aside from the carbohydrate attached to the heavy chain, up to 30% of human IgGs have a glycosylated Fab region. IgG has a single N-linked biantennary carbohydrate at Asn297 of the CH2 domain. For IgG from either serum or produced ex vivo in hybridomas or engineered cells, the IgG are heterogeneous with respect to the Asn297 linked
carbohydrate. For human IgG, the core oligosaccharide normally consists of
GlcNAc2Man3GlcNAc, with differing numbers of outer residues.
[161] The carbohydrate moieties of immunoglobulins disclosed herein will be described with reference to commonly used nomenclature for the description of oligosaccharides. A review of carbohydrate chemistry which uses this nomenclature is found in Hubbard et al. 1981 , Ann. Rev. Biochem. 50:555-583. This nomenclature includes, for instance, Man, which represents mannose; GlcNAc, which represents 2-N-acetylglucosamine; Gal which represents galactose; Fuc for fucose; and Glc, which represents glucose. Sialic acids are described by the shorthand notation NeuNAc, for 5-N-acetylneuraminic acid, and NeuNGc for 5-glycolylneuraminic.
[162] The term "glycosylation" means the attachment of oligosaccharides (carbohydrates containing two or more simple sugars linked together e.g. from two to about twelve simple sugars linked together) to a glycoprotein. The oligosaccharide side chains are typically linked to the backbone of the glycoprotein through either N- or O-linkages. The oligosaccharides of immunoglobulins disclosed herein occur generally are attached to a CH2 domain of an Fc region as N-linked oligosaccharides. "N-linked glycosylation" refers to the attachment of the carbohydrate moiety to an asparagine residue in a glycoprotein chain. The skilled artisan will recognize that, for example, each of murine lgG1 , lgG2a, lgG2b and lgG3 as well as human lgG1 , lgG2, lgG3, lgG4, IgA and IgD CH2 domains have a single site for N-linked
glycosylation at residue 297.
[163] For the purposes herein, a "mature core carbohydrate structure" refers to a processed core carbohydrate structure attached to an Fc region which generally consists of the following carbohydrate structure GlcNAc(Fucose)-GlcNAc-Man-(Man-GlcNAc)2 typical of biantennary oligosaccharides. The mature core carbohydrate structure is attached to the Fc region of the glycoprotein, generally via N-linkage to Asn297 of a CH2 domain of the Fc region. A "bisecting GlcNAc" is a GlcNAc residue attached to the a1 ,4 mannose of the mature core carbohydrate structure. The bisecting GlcNAc can be enzymatically attached to the mature core carbohydrate structure by a a(1 ,4)-N-acetylglucosaminyltransferase III enzyme (GnTIII). CHO cells do not normally express GnTIII (Stanley et al., 1984, J. Biol. Chem. 261 :13370-13378), but may be engineered to do so (Umana et al., 1999, Nature Biotech. 17:176-180).
[164] Described herein are immunoglobulins that comprise modified glycoforms or engineered glycoforms. By "modified glycoform" or "engineered glycoform" as used herein is meant a carbohydrate composition that is covalently attached to a protein, for example an antibody, wherein said carbohydrate composition differs chemically from that of a parent protein. Engineered glycoforms may be useful for a variety of purposes, including but not limited to enhancing or reducing FcyR-mediated effector function. In one embodiment, the immunoglobulins disclosed herein are modified to control the level of fucosylated and/or bisecting oligosaccharides that are covalently attached to the Fc region.
[165] A variety of methods are well known in the art for generating modified glycoforms (Umana et al., 1999, Nat Biotechnol 17:176-180; Davies et al., 2001 , Biotechnol Bioeng 74:288-294; Shields et al., 2002, J Biol Chem 277:26733-26740; Shinkawa et al., 2003, J Biol Chem 278:3466-3473; USSN 12/434,533; all of which are expressly incorporated by reference). These techniques control the level of fucosylated and/or bisecting
oligosaccharides that are covalently attached to the Fc region, for example by expressing an IgG in various organisms or cell lines, engineered or otherwise (for example Lec-13 CHO cells or rat hybridoma YB2/0 cells), by regulating enzymes involved in the glycosylation pathway (for example FUT8 [a1 ,6-fucosyltranserase] and/or β1-4- N- acetylglucosaminyltransferase III [GnTIII]), by modifying carbohydrate(s) after the IgG has been expressed, or by expressing antibody in the presence of fucose analogs as enzymatic inhibitors. Other methods for modifying glycoforms of the immunoglobulins disclosed herein include using glycoengineered strains of yeast (Li et al., 2006, Nature Biotechnology
24(2):210-215), moss (Nechansky et al., 2007, Mol Immunjol 44(7): 1826-8), and plants (Cox et al., 2006, Nat Biotechnol 24(12):1591 -7). The use of a particular method to generate a modified glycoform is not meant to constrain embodiments to that method. Rather, embodiments disclosed herein encompass immunoglobulins with modified glycoforms irrespective of how they are produced.
[166] In one embodiment, immunoglobulins disclosed herein are glycoengineered to alter the level of sialylation. Higher levels of sialylated Fc glycans in immunoglobulin G molecules can adversely impact functionality (Scallon et al., 2007, Mol Immunol. 44(7):1524-34), and differences in levels of Fc sialylation can result in modified anti-inflammatory activity (Kaneko et al., 2006, Science 313:670-673). Because antibodies may acquire anti-inflammatory properties upon sialylation of Fc core polysaccharide, it may be advantageous to
glycoengineer the immunoglobulins disclosed herein for greater or reduced Fc sialic acid content.
[167] Engineered glycoform typically refers to the different carbohydrate or
oligosaccharide; thus for example an immuoglobulin may comprise an engineered glycoform. Alternatively, engineered glycoform may refer to the immunoglobulin that comprises the different carbohydrate or oligosaccharide. In one embodiment, a composition disclosed herein comprises a glycosylated immunoglobulin having an Fc region, wherein about 51-100% of the glycosylated antibody, e.g., 80-100%, 90-100%, 95-100%, etc. of the antibody in the composition comprises a mature core carbohydrate structure which lacks fucose. In another embodiment, the antibody in the composition both comprises a mature core carbohydrate structure that lacks fucose and additionally comprises at least one amino acid modification in the Fc region. In an alternative embodiment, a composition comprises a glycosylated immunoglobulin having an Fc region, wherein about 51-100% of the
glycosylated antibody, 80-100%, or 90-100%, of the antibody in the composition comprises a mature core carbohydrate structure which lacks sialic acid. In another embodiment, the antibody in the composition both comprises a mature core carbohydrate structure that lacks sialic acid and additionally comprises at least one amino acid modification in the Fc region. In yet another embodiment, a composition comprises a glycosylated immunoglobulin having an Fc region, wherein about 51 -100% of the glycosylated antibody, 80-100%, or 90-100%, of the antibody in the composition comprises a mature core carbohydrate structure which contains sialic acid. In another embodiment, the antibody in the composition both comprises a mature core carbohydrate structure that contains sialic acid and additionally comprises at least one amino acid modification in the Fc region. In another embodiment, the combination of engineered glycoform and amino acid modification provides optimal Fc receptor binding properties to the antibody.
[168] Other Modifications [169] Immunoglobulins disclosed herein may comprise one or more modifications that provide optimized properties. Said modifications may be amino acid modifications, or may be modifications that are made enzymatically or chemically. Such modification(s) likely provide some improvement in the immunoglobulin, for example an enhancement in its stability, solubility, function, or clinical use. Disclosed herein are a variety of improvements that may be made by coupling the immunoglobulins disclosed herein with additional modifications.
[170] In one embodiment, the variable region of an immunoglobulin disclosed herein may be affinity matured, that is to say that amino acid modifications have been made in the VH and/or VL domains to enhance binding of the antibody to its target antigen. Such types of modifications may improve the association and/or the dissociation kinetics for binding to the target antigen. Other modifications include those that improve selectivity for target antigen vs. alternative targets. These include modifications that improve selectivity for antigen expressed on target vs. non-target cells. Immunoglobulins disclosed herein may comprise one or more modifications that provide reduced or enhanced internalization of an
immunoglobulin.
[171] In one embodiment, modifications are made to improve biophysical properties of the immunoglobulins disclosed herein, including but not limited to stability, solubility, and oligomeric state. Modifications can include, for example, substitutions that provide more favorable intramolecular interactions in the immunoglobulin such as to provide greater stability, or substitution of exposed nonpolar amino acids with polar amino acids for higher solubility. Other modifications to the immunoglobulins disclosed herein include those that enable the specific formation or homodimeric or homomultimeric molecules. Such modifications include but are not limited to engineered disulfides, as well as chemical modifications or aggregation methods.
[172] In further embodiments, the immunoglobulins disclosed herein comprise
modifications that remove proteolytic degradation sites. These may include, for example, protease sites that reduce production yields, as well as protease sites that degrade the administered protein in vivo. In one embodiment, additional modifications are made to remove covalent degradation sites such as deamidation (i.e. deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues), oxidation, and proteolytic degradation sites. Deamidation sites that are particular useful to remove are those that have enhance propensity for deamidation, including, but not limited to asparaginyl and gltuamyl residues followed by glycines (NG and QG motifs, respectively). In such cases, substitution of either residue can significantly reduce the tendency for deamidation. Common oxidation sites include methionine and cysteine residues. Other covalent modifications, that can either be introduced or removed, include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the "-amino groups of lysine, arginine, and histidine side chains, acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group. Additional modifications also may include but are not limited to posttranslational modifications such as N-linked or O-linked glycosylation and phosphorylation.
[173] Modifications may include those that improve expression and/or purification yields from hosts or host cells commonly used for production of biologies. These include, but are not limited to various mammalian cell lines (e.g. CHO), yeast cell lines, bacterial cell lines, and plants. Additional modifications include modifications that remove or reduce the ability of heavy chains to form inter-chain disulfide linkages. Additional modifications include modifications that remove or reduce the ability of heavy chains to form intra-chain disulfide linkages.
[174] The immunoglobulins disclosed herein may comprise modifications that include the use of unnatural amino acids incorporated using, including but not limited to methods described in Liu & Schultz, 2010, Annu Rev Biochem 79:413-444, herein expressly incorporated by reference. In some embodiments, these modifications enable manipulation of various functional, biophysical, immunological, or manufacturing properties discussed above. In additional embodiments, these modifications enable additional chemical modification for other purposes.
[175] Other modifications are contemplated herein. For example, the immunoglobulin may be linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and
polypropylene glycol. Additional amino acid modifications may be made to enable specific or non-specific chemical or posttranslational modification of the immunoglobulins. Such modifications, include, but are not limited to PEGylation and glycosylation. Specific substitutions that can be utilized to enable PEGylation include, but are not limited to, introduction of novel cysteine residues or unnatural amino acids such that efficient and specific coupling chemistries can be used to attach a PEG or otherwise polymeric moiety. Introduction of specific glycosylation sites can be achieved by introducing novel N-X-T/S sequences into the immunoglobulins disclosed herein.
[176] Modifications to reduce immunogenicity may include modifications that reduce binding of processed peptides derived from the parent sequence to MHC proteins. For example, amino acid modifications would be engineered such that there are no or a minimal number of immune epitopes that are predicted to bind, with high affinity, to any prevalent MHC alleles. Several methods of identifying MHC-binding epitopes in protein sequences are known in the art and may be used to score epitopes in an antibody disclosed herein.
[177] Covalent modifications are included within the scope of immunoglobulins disclosed herein, and are generally, but not always, done post-translationally. For example, several types of covalent modifications can be introduced into the molecule by reacting specific amino acid residues with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues. In some embodiments, the covalent modification of the immunogloublins disclosed herein comprises the addition of one or more labels. The term "labeling group" means any detectable label. In some embodiments, the labeling group is coupled to the immunoglobulin via spacer arms of various lengths to reduce potential steric hindrance. Various methods for labeling proteins are known in the art and may be used in generating immunoglobulins disclosed herein.
[178] Conjugates
[179] In one embodiment, the immunoglobulins disclosed herein are "fusion proteins", sometimes referred to herein as "conjugates". The fusion partner or conjugate partner can be proteinaceous or non-proteinaceous; the latter generally being generated using functional groups on the immunoglobulin and on the conjugate partner. Conjugate and fusion partners may be any molecule, including small molecule chemical compounds and polypeptides. For example, a variety of conjugates and methods are described in Trail et al., 1999, Curr. Opin. Immunol. 1 1 :584-588, incorporated entirely by reference. Possible conjugate partners include but are not limited to cytokines, cytotoxic agents, toxins, radioisotopes,
chemotherapeutic agent, anti-angiogenic agents, a tyrosine kinase inhibitors, and other therapeutically active agents. In some embodiments, conjugate partners may be thought of more as payloads, that is to say that the goal of a conjugate is targeted delivery of the conjugate partner to a targeted cell, for example a cancer cell or immune cell, by the immunoglobulin. Thus, for example, the conjugation of a toxin to an immunoglobulin targets the delivery of said toxin to cells expressing the target antigen. As will be appreciated by one skilled in the art, in reality the concepts and definitions of fusion and conjugate are overlapping. The designation of a fusion or conjugate is not meant to constrain it to any particular embodiment disclosed herein. Rather, these terms are used loosely to convey the broad concept that any immunoglobulin disclosed herein may be linked genetically, chemically, or otherwise, to one or more polypeptides or molecules to provide some desirable property.
[180] Suitable conjugates include, but are not limited to, labels as described below, drugs and cytotoxic agents including, but not limited to, cytotoxic drugs (e.g., chemotherapeutic agents) or toxins or active fragments of such toxins. Suitable toxins and their corresponding fragments include diptheria A chain, exotoxin A chain, ricin A chain, abrin A chain, curcin, crotin, phenomycin, enomycin and the like. Cytotoxic agents also include radiochemicals made by conjugating radioisotopes to immunoglobulin, or binding of a radionuclide to a chelating agent that has been covalently attached to the immunoglobulin. Additional embodiments utilize calicheamicin, auristatins, geldanamycin, maytansine, and
duocarmycins and analogs. Antibody-drug conjugates are described in Alley et al., 2010, Curr Opin Chem Biol 14[4]:529-37, herein expreslly incorporated by reference.
[181] In one embodiment, the immunoglobulins disclosed herein are fused or conjugated to a cytokine. By "cytokine" as used herein is meant a generic term for proteins released by one cell population that act on another cell as intercellular mediators. For example, as described in Penichet et al., 2001 , J. Immunol. Methods 248:91-101 , incorporated entirely by reference, cytokines may be fused to an immunoglobulin to provide an array of desirable properties. Examples of such cytokines are lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone;
parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin;
placental lactogen; tumor necrosis factor-alpha and -beta; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-beta; platelet-growth factor; transforming growth factors (TGFs) such as TGF-alpha and TGF-beta; insulin-like growth factor-l and -II; erythropoietin (EPO); osteoinductive factors; interferons such as interferon-alpha, beta, and -gamma; colony stimulating factors (CSFs) such as macrophage- CSF (M-CSF); granulocyte-macrophage-CSF (GM-CSF); and granulocyte-CSF (G-CSF); interleukins (ILs) such as IL-1 , IL-1 alpha, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL- 1 1 , IL-12; IL-15, a tumor necrosis factor such as TNF-alpha or TNF-beta; C5a; and other polypeptide factors including LIF and kit ligand (KL). As used herein, the term cytokine includes proteins from natural sources or from recombinant cell culture, and biologically active equivalents of the native sequence cytokines.
[182] In yet another embodiment, an immunoglobulins disclosed herein may be conjugated to a "receptor" (such streptavidin) for utilization in tumor pretargeting wherein the
immunoglobulin-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g. avidin) which is conjugated to a cytotoxic agent (e.g. a radionucleotide). In an alternate embodiment, the immunoglobulin is conjugated or operably linked to an enzyme in order to employ Antibody Dependent Enzyme Mediated Prodrug Therapy (ADEPT). ADEPT may be used by conjugating or operably linking the immunoglobulin to a prodrug-activating enzyme that converts a prodrug (e.g. a peptidyl chemotherapeutic agent.
[183] Production of Immunoglobulins
[184] Also disclosed herein are methods for producing and experimentally testing immunoglobulins. The disclosed methods are not meant to constrain embodiments to any particular application or theory of operation. Rather, the provided methods are meant to illustrate generally that one or more immunoglobulins may be produced and experimentally tested to obtain immunoglobulins. General methods for antibody molecular biology, expression, purification, and screening are described in Antibody Engineering, edited by Kontermann & Dubel, Springer, Heidelberg, 2001 ; and Hayhurst & Georgiou, 2001 , Curr Opin Chem Biol 5:683-689; Maynard & Georgiou, 2000, Annu Rev Biomed Eng 2:339-76;
[185] In one embodiment disclosed herein, nucleic acids are created that encode the immunoglobulins, and that may then be cloned into host cells, expressed and assayed, if desired. Thus, nucleic acids, and particularly DNA, may be made that encode each protein sequence. These practices are carried out using well-known procedures. For example, a variety of methods that may find use in generating immunoglobulins disclosed herein are described in Molecular Cloning - A Laboratory Manual, 3rd Ed. (Maniatis, Cold Spring Harbor Laboratory Press, New York, 2001 ), and Current Protocols in Molecular Biology (John Wiley & Sons), both incorporated entirely by reference. There are a variety of techniques that may be used to efficiently generate DNA encoding immunoglobulins disclosed herein. Such methods include but are not limited to gene assembly methods, PCR-based method and methods which use variations of PCR, ligase chain reaction-based methods, pooled oligo methods such as those used in synthetic shuffling, error-prone amplification methods and methods which use oligos with random mutations, classical site- directed mutagenesis methods, cassette mutagenesis, and other amplification and gene synthesis methods. As is known in the art, there are a variety of commercially available kits and methods for gene assembly, mutagenesis, vector subcloning, and the like, and such commercial products find use in for generating nucleic acids that encode immunoglobulins.
[186] The immunoglobulin proteins disclosed herein may be produced by culturing a host cell transformed with nucleic acid, e.g., an expression vector, containing nucleic acid encoding the immunoglobulins, under the appropriate conditions to induce or cause expression of the protein. The conditions appropriate for expression will vary with the choice of the expression vector and the host cell, and will be easily ascertained by one skilled in the art through routine experimentation. A wide variety of appropriate host cells may be used, including but not limited to mammalian cells, bacteria, insect cells, yeast, and plant cells. For example, a variety of cell lines that may find use in generating immunoglobulins disclosed herein are described in the ATCC® cell line catalog, available from the American Type Culture Collection.
[187] In one embodiment, the immunoglobulins are expressed in mammalian expression systems, including systems in which the expression constructs are introduced into the mammalian cells using virus such as retrovirus or adenovirus. Any mammalian cells may be used, e.g., human, mouse, rat, hamster, and primate cells. Suitable cells also include known research cells, including but not limited to Jurkat T cells, NIH3T3, CHO, BHK, COS,
HEK293, PER C.6, HeLa, Sp2/0, NSO cells and variants thereof. In an alternateembodiment, library proteins are expressed in bacterial cells. Bacterial expression systems are well known in the art, and include Escherichia coli (E. coli), Bacillus subtilis, Streptococcus cremoris, and Streptococcus lividans. In alternate embodiments, immunoglobulins are produced in insect cells (e.g. Sf21/Sf9, Trichoplusia ni Bti-Tn5b1 -4) or yeast cells (e.g. S. cerevisiae, Pichia, etc). In an alternate embodiment, immunoglobulins are expressed in vitro using cell free translation systems. In vitro translation systems derived from both prokaryotic (e.g. E. coli) and eukaryotic (e.g. wheat germ, rabbit reticulocytes) cells are available and may be chosen based on the expression levels and functional properties of the protein of interest. For example, as appreciated by those skilled in the art, in vitro translation is required for some display technologies, for example ribosome display. In addition, the immunoglobulins may be produced by chemical synthesis methods. Also transgenic expression systems both animal (e.g. cow, sheep or goat milk, embryonated hen's eggs, whole insect larvae, etc.) and plant (e.g. corn, tobacco, duckweed, etc.)
[188] The nucleic acids that encode the immunoglobulins disclosed herein may be incorporated into an expression vector in order to express the protein. A variety of expression vectors may be utilized for protein expression. Expression vectors may comprise self-replicating extra-chromosomal vectors or vectors which integrate into a host genome. Expression vectors are constructed to be compatible with the host cell type. Thus expression vectors which find use in generating immunoglobulins disclosed herein include but are not limited to those which enable protein expression in mammalian cells, bacteria, insect cells, yeast, and in in vitro systems. As is known in the art, a variety of expression vectors are available, commercially or otherwise, that may find use for expressing immunoglobulins disclosed herein.
[189] Expression vectors typically comprise a protein operably linked with control or regulatory sequences, selectable markers, any fusion partners, and/or additional elements. By "operably linked" herein is meant that the nucleic acid is placed into a functional relationship with another nucleic acid sequence. Generally, these expression vectors include transcriptional and translational regulatory nucleic acid operably linked to the nucleic acid encoding the immunoglobulin, and are typically appropriate to the host cell used to express the protein. In general, the transcriptional and translational regulatory sequences may include promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences. As is also known in the art, expression vectors typically contain a selection gene or marker to allow the selection of transformed host cells containing the expression vector. Selection genes are well known in the art and will vary with the host cell used.
[190] Immunoglobulins may be operably linked to a fusion partner to enable targeting of the expressed protein, purification, screening, display, and the like. Fusion partners may be linked to the immunoglobulin sequence via a linker sequences. The linker sequence will generally comprise a small number of amino acids, typically less than ten, although longer linkers may also be used. Typically, linker sequences are selected to be flexible and resistant to degradation. As will be appreciated by those skilled in the art, any of a wide variety of sequences may be used as linkers. For example, a common linker sequence comprises the amino acid sequence GGGGS. A fusion partner may be a targeting or signal sequence that directs immunoglobulin and any associated fusion partners to a desired cellular location or to the extracellular media. As is known in the art, certain signaling sequences may target a protein to be either secreted into the growth media, or into the periplasmic space, located between the inner and outer membrane of the cell. A fusion partner may also be a sequence that encodes a peptide or protein that enables purification and/or screening. Such fusion partners include but are not limited to polyhistidine tags (His- tags) (for example H6 and H 10 or other tags for use with Immobilized Metal Affinity
Chromatography (IMAC) systems (e.g. Ni+2 affinity columns)), GST fusions, MBP fusions, Strep-tag, the BSP biotinylation target sequence of the bacterial enzyme BirA, and epitope tags which are targeted by antibodies (for example c-myc tags, flag-tags, and the like). As will be appreciated by those skilled in the art, such tags may be useful for purification, for screening, or both. For example, an immunoglobulin may be purified using a His-tag by immobilizing it to a Ni+2 affinity column, and then after purification the same His-tag may be used to immobilize the antibody to a Ni+2 coated plate to perform an ELISA or other binding assay (as described below). A fusion partner may enable the use of a selection method to screen immunoglobulins (see below). Fusion partners that enable a variety of selection methods are well-known in the art. [191] For example, by fusing the members of an immunoglobulin library to the gene III protein, phage display can be employed. Fusion partners may enable immunoglobulins to be labeled. Alternatively, a fusion partner may bind to a specific sequence on the expression vector, enabling the fusion partner and associated immunoglobulin to be linked covalently or noncovalently with the nucleic acid that encodes them. The methods of introducing exogenous nucleic acid into host cells are well known in the art, and will vary with the host cell used. Techniques include but are not limited to dextran-mediated transfection, calcium phosphate precipitation, calcium chloride treatment, polybrene mediated transfection, protoplast fusion, electroporation, viral or phage infection, encapsulation of the
polynucleotide(s) in liposomes, and direct microinjection of the DNA into nuclei. In the case of mammalian cells, transfection may be either transient or stable.
[192] In one embodiment, immunoglobulins are purified or isolated after expression.
Proteins may be isolated or purified in a variety of ways known to those skilled in the art. Purification may be particularly useful in the invention for separating heterodimeric heavy chain species from homodimeric heavy chain species, as described herein. Standard purification methods include chromatographic techniques, including ion exchange, hydrophobic interaction, affinity, sizing or gel filtration, and reversed-phase, carried out at atmospheric pressure or at high pressure using systems such as FPLC and HPLC.
Purification methods also include electrophoretic, isoelectric focusing, immunological, precipitation, dialysis, and chromatofocusing techniques. Ultrafiltration and diafiltration techniques, in conjunction with protein concentration, are also useful. As is well known in the art, a variety of natural proteins bind Fc and antibodies, and these proteins can find use for purification of immunoglobulins disclosed herein. For example, the bacterial proteins A and G bind to the Fc region. Likewise, the bacterial protein L binds to the Fab region of some antibodies, as of course does the antibody's target antigen. Purification can often be enabled by a particular fusion partner. For example, immunoglobulins may be purified using glutathione resin if a GST fusion is employed, Ni+2 affinity chromatography if a His-tag is employed, or immobilized anti-flag antibody if a flag-tag is used. For general guidance in suitable purification techniques, see, e.g. incorporated entirely by reference Protein
Purification: Principles and Practice, 3rd Ed., Scopes, Springer-Verlag, NY, 1994, incorporated entirely by reference. The degree of purification necessary will vary depending on the screen or use of the immunoglobulins. In some instances no purification is necessary. For example in one embodiment, if the immunoglobulins are secreted, screening may take place directly from the media. As is well known in the art, some methods of selection do not involve purification of proteins.
[193] In Vitro Experimentation [194] Immunoglobulins may be screened using a variety of in vitro methods, including but not limited to those that use binding assays, cell-based assays, and selection technologies. Automation and high-throughput screening technologies may be utilized in the screening procedures. Screening may employ the use of a fusion partner or label. The use of fusion partners has been discussed above. By "labeled" herein is meant that the immunoglobulins disclosed herein have one or more elements, isotopes, or chemical compounds attached to enable the detection in a screen. In general, labels fall into three classes: a) immune labels, which may be an epitope incorporated as a fusion partner that is recognized by an antibody, b) isotopic labels, which may be radioactive or heavy isotopes, and c) small molecule labels, which may include fluorescent and colorimetric dyes, or molecules such as biotin that enable other labeling methods. Labels may be incorporated into the compound at any position and may be incorporated in vitro or in vivo during protein expression.
[195] In one embodiment, the functional and/or biophysical properties of immunoglobulins are screened in an in vitro assay. In vitro assays may allow a broad dynamic range for screening properties of interest. Particularly relevant for the present invention, the
immunoglobulins may be tested for their affinity for one or more antigens. Properties that may be screened include but are not limited to stability, solubility, and affinity for Fc ligands, for example FcyRs. Multiple properties may be screened simultaneously or individually.
Proteins may be purified or unpurified, depending on the requirements of the assay. In one embodiment, the screen is a qualitative or quantitative binding assay for binding of immunoglobulins to a protein or nonprotein molecule that is known or thought to bind the immunoglobulin. In one embodiment, the screen is a binding assay for measuring binding to the target antigen. In an alternate embodiment, the screen is an assay for binding of immunoglobulins to an Fc ligand, including but are not limited to the family of FcyRs, the neonatal receptor FcRn, the complement protein C1 q, and the bacterial proteins A and G. Said Fc ligands may be from any organism. In one embodiment, Fc ligands are from humans, mice, rats, rabbits, and/or monkeys. Binding assays can be carried out using a variety of methods known in the art, including but not limited to FRET (Fluorescence
Resonance Energy Transfer) and BRET (Bioluminescence Resonance Energy Transfer) - based assays, AlphaScreen™ (Amplified Luminescent Proximity Homogeneous Assay), Scintillation Proximity Assay, ELISA (Enzyme-Linked Immunosorbent Assay), SPR (Surface Plasmon Resonance, also known as BIACORE®), isothermal titration calorimetry, differential scanning calorimetry, gel electrophoresis, and chromatography including gel filtration. These and other methods may take advantage of some fusion partner or label of the
immunoglobulin. Assays may employ a variety of detection methods including but not limited to chromogenic, fluorescent, luminescent, or isotopic labels. [196] The biophysical properties of immunoglobulins, for example stability and solubility, may be tested using a variety of methods known in the art. Protein stability may be determined by measuring the thermodynamic equilibrium between folded and unfolded states. For example, immunoglobulins disclosed herein may be unfolded using chemical denaturant, heat, or pH, and this transition may be monitored using methods including but not limited to circular dichroism spectroscopy, fluorescence spectroscopy, absorbance spectroscopy, NMR spectroscopy, calorimetry, and proteolysis. As will be appreciated by those skilled in the art, the kinetic parameters of the folding and unfolding transitions may also be monitored using these and other techniques. The solubility and overall structural integrity of an immunoglobulin may be quantitatively or qualitatively determined using a wide range of methods that are known in the art. Methods which may find use for characterizing the biophysical properties of immunoglobulins disclosed herein include gel electrophoresis, isoelectric focusing, capillary electrophoresis, chromatography such as size exclusion chromatography, ion-exchange chromatography, and reversed-phase high performance liquid chromatography, peptide mapping, oligosaccharide mapping, mass spectrometry, ultraviolet absorbance spectroscopy, fluorescence spectroscopy, circular dichroism spectroscopy, isothermal titration calorimetry, differential scanning calorimetry, analytical ultra-centrifugation, dynamic light scattering, proteolysis, and cross-linking, turbidity measurement, filter retardation assays, immunological assays, fluorescent dye binding assays, protein-staining assays, microscopy, and detection of aggregates via ELISA or other binding assay. Structural analysis employing X-ray crystallographic techniques and NMR spectroscopy may also find use. In one embodiment, stability and/or solubility may be measured by determining the amount of protein solution after some defined period of time. In this assay, the protein may or may not be exposed to some extreme condition, for example elevated temperature, low pH, or the presence of denaturant. Because function typically requires a stable, soluble, and/or well-folded/structured protein, the aforementioned functional and binding assays also provide ways to perform such a measurement. For example, a solution comprising an immunoglobulin could be assayed for its ability to bind target antigen, then exposed to elevated temperature for one or more defined periods of time, then assayed for antigen binding again. Because unfolded and aggregated protein is not expected to be capable of binding antigen, the amount of activity remaining provides a measure of the immunoglobulin's stability and solubility.
[197] In one embodiment, immunoglobulins may be tested using one or more cell-based or in vitro assays. For such assays, immunoglobulins, purified or unpurified, are typically added exogenously such that cells are exposed to immunoglobulins described herein. These assays are typically, but not always, based on the biology of the ability of the immunoglobulin to bind to the target antigen and mediate some biochemical event, for example effector functions like cellular lysis, phagocytosis, ligand/receptor binding inhibition, inhibition of growth and/or proliferation, inhibition of calcium release and/or signaling, apoptosis and the like. Such assays often involve monitoring the response of cells to immunoglobulin, for example cell survival, cell death, cellular phagocytosis, cell lysis, change in cellular morphology, or transcriptional activation such as cellular expression of a natural gene or reporter gene. For example, such assays may measure the ability of
immunoglobulins to elicit cell killing, for example ADCC, ADCP, and CDC. Assays that measure cellular killing that is mediated by co-engagement of antigens are particularly relevant for the invention. For some assays additional cells or components, that is in addition to the target cells, may need to be added, for example serum complement, or effector cells such as peripheral blood monocytes (PBMCs), NK cells, macrophages, T cells, and the like. Such additional cells may be from any organism, e.g., humans, mice, rat, rabbit, and monkey. Crosslinked or monomeric antibodies may cause apoptosis of certain cell lines expressing the antibody's target antigen, or they may mediate attack on target cells by immune cells which have been added to the assay. Methods for monitoring cell death or viability are known in the art, and include the use of dyes, fluorophores, immunochemical, cytochemical, and radioactive reagents. For example, caspase assays or annexin- flourconjugates may enable apoptosis to be measured, and uptake or release of radioactive substrates (e.g. Chromium-51 release assays) or the metabolic reduction of fluorescent dyes such as alamar blue may enable cell growth, proliferation or activation to be monitored. In one embodiment, the DELFIA EuTDA-based cytotoxicity assay (Perkin Elmer, MA) is used. Alternatively, dead or damaged target cells may be monitored by measuring the release of one or more natural intracellular proteins, for example lactate dehydrogenase.
Transcriptional activation may also serve as a method for assaying function in cell-based assays. In this case, response may be monitored by assaying for natural genes or proteins which may be upregulated or down-regulated, for example the release of certain interleukins may be measured, or alternatively readout may be via a luciferase or GFP-reporter construct. Cell-based assays may also involve the measure of morphological changes of cells as a response to the presence of an immunoglobulin. Cell types for such assays may be prokaryotic or eukaryotic, and a variety of cell lines that are known in the art may be employed. Alternatively, cell-based screens are performed using cells that have been transformed or transfected with nucleic acids encoding the immunoglobulins.
[198] In Vivo Experimentation
[199] The biological properties of the immunoglobulins disclosed herein may be
characterized in cell, tissue, and whole organism experiments. As is known in the art, drugs are often tested in animals, including but not limited to mice, rats, rabbits, dogs, cats, pigs, and monkeys, in order to measure a drug's efficacy for treatment against a disease or disease model, or to measure a drug's pharmacokinetics, toxicity, and other properties. Said animals may be referred to as disease models. With respect to the immunoglobulins disclosed herein, a particular challenge arises when using animal models to evaluate the potential for in-human efficacy of candidate polypeptides - this is due, at least in part, to the fact that immunoglobulins that have a specific effect on the affinity for a human Fc receptor may not have a similar affinity effect with the orthologous animal receptor. These problems can be further exacerbated by the inevitable ambiguities associated with correct assignment of true orthologues (Mechetina et al., 2002, Immunogenetics 54:463-468, incorporated entirely by reference), and the fact that some orthologues simply do not exist in the animal. Therapeutics are often tested in mice, including but not limited to nude mice, Rag-deficient mice, SCID mice, xenograft mice, and transgenic mice (including knockins and knockouts). For example, an immunoglobulin of the present invention that is intended as an anti-cancer therapeutic may be tested in a mouse cancer model, for example a xenograft mouse. In this method, a tumor or tumor cell line is grafted onto or injected into a mouse, and subsequently the mouse is treated with the therapeutic to determine the ability of the drug to reduce or inhibit cancer growth and metastasis. Therapeutic immunoglobulins herein can be tested in mouse strains NZB, NOD, BXSB, MRL/lpr, K/BxN and transgenics (including knockins and knockouts). Such mice can develop various autoimmune conditions that resemble human organ specific, systemic autoimmune or inflammatory disease pathologies such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). For example, an immunoglobulin disclosed herein intended for autoimmune diseases may be tested in such mouse models by treating the mice to determine the ability of the immunoglobulin to reduce or inhibit the development of the disease pathology. Because of the incompatibility between the mouse and human Fey receptor system, an alternative approach is to use a murine SCID model in which immune deficient mice are engrafted with human PBLs or PBMCs (huPBL-SCID, huPBMC-SCID) providing a semi-functional human immune system with human effector cells and Fc receptors. Other organisms, e.g., mammals, may also be used for testing. For example, because of their genetic similarity to humans, monkeys can be suitable therapeutic models, and thus may be used to test the efficacy, toxicity, pharmacokinetics, or other property of the immunoglobulins disclosed herein. Tests of the immunoglobulins disclosed herein in humans are ultimately required for approval as drugs, and thus of course these experiments are contemplated. Thus the immunoglobulins disclosed herein may be tested in humans to determine their therapeutic efficacy, toxicity, pharmacokinetics, and/or other clinical properties. [200] In some embodiments, immunoglobulins disclosed herein may be assessed for efficacy in clinically relevant animal models of various human diseases. In many cases, relevant models include various transgenic animals for specific antigens and receptors.
[201] In one embodiment, the testing of immunoglobulins may include study of efficacy in primates (e.g. cynomolgus monkey model) to facilitate the evaluation of depletion of specific target cells harboring the target antigen. Additional primate models include but are not limited to use of the rhesus monkey to assess immunoglobulins in therapeutic studies of autoimmune, transplantation and cancer.
[202] Toxicity studies are performed to determine drug related-effects that cannot be evaluated in standard pharmacology profiles, or occur only after repeated administration of the agent. Most toxicity tests are performed in two species - a rodent and a non-rodent - to ensure that any unexpected adverse effects are not overlooked before new therapeutic entities are introduced into man. In general, these models may measure a variety of toxicities including genotoxicity, chronic toxicity, immunogenicity,
reproductive/developmental toxicity and carcinogenicity. Included within the aforementioned parameters are standard measurement of food consumption, bodyweight, antibody formation, clinical chemistry, and macro- and microscopic examination of standard organs/tissues (e.g. cardiotoxicity). Additional parameters of measurement are injection site trauma and the measurement of neutralizing antibodies, if any. Traditionally, monoclonal antibody therapeutics, naked or conjugated, is evaluated for cross-reactivity with normal tissues, immunogenicity/antibody production, conjugate or linker toxicity and "bystander" toxicity of radiolabeled species. Nonetheless, such studies may have to be individualized to address specific concerns and following the guidance set by ICH S6 (Safety studies for biotechnological products, also noted above). As such, the general principles are that the products are sufficiently well characterized, impurities/contaminants have been removed, that the test material is comparable throughout development, and that GLP compliance is maintained.
[203] The pharmacokinetics (PK) of the immunoglobulins disclosed herein may be studied in a variety of animal systems, with the most relevant being non-human primates such as the cynomolgus and rhesus monkeys. Single or repeated i.v./s.c. administrations over a dose range of 6000-fold (0.05-300 mg/kg) can be evaluated for half-life (days to weeks) using plasma concentration and clearance. Volume of distribution at a steady state and level of systemic absorbance can also be measured. Examples of such parameters of measurement generally include maximum observed plasma concentration (Cmax), the time to reach Cmax (Tmax), the area under the plasma concentration-time curve from time 0 to infinity [AUC(0- inf] and apparent elimination half-life (T1 /2). Additional measured parameters could include compartmental analysis of concentration-time data obtained following i.v. administration and bioavailability.
[204] Pharmacodynamic studies may include, but are not limited to, targeting specific cells or blocking signaling mechanisms, measuring inhibition of antigen-specific antibodies etc. The immunoglobulins disclosed herein may target particular effector cell populations and thereby be direct drugs to induce certain activities to improve potency or to increase penetration into a particularly favorable physiological compartment. Such pharmacodynamic effects may be demonstrated in animal models or in humans.
[205] Clinical Use
[206] The immunoglobulins disclosed herein may find use in a wide range of products. In one embodiment an immunoglobulin disclosed herein is a therapeutic, a diagnostic, or a research reagent. The immunoglobulins may find use in a composition that is monoclonal or polyclonal. The immunoglobulins disclosed herein may be used for therapeutic purposes. As will be appreciated by those in the art, the immunoglobulins disclosed herein may be used for any therapeutic purpose that antibodies, Fc fusions, and the like may be used for. The immunoglobulins may be administered to a patient to treat disorders including but not limited to cancer, infectious diseases, autoimmune and inflammatory diseases.
[207] A "patient" for the purposes disclosed herein includes both humans and other animals, e.g., other mammals. Thus the immunoglobulins disclosed herein have both human therapy and veterinary applications. The term "treatment" or "treating" as disclosed herein is meant to include therapeutic treatment, as well as prophylactic, or suppressive measures for a disease or disorder. Thus, for example, successful administration of an immunoglobulin prior to onset of the disease results in treatment of the disease. As another example, successful administration of an optimized immunoglobulin after clinical manifestation of the disease to combat the symptoms of the disease comprises treatment of the disease.
"Treatment" and "treating" also encompasses administration of an optimized immunoglobulin after the appearance of the disease in order to eradicate the disease. Successful administration of an agent after onset and after clinical symptoms have developed, with possible abatement of clinical symptoms and perhaps amelioration of the disease, comprises treatment of the disease. Those "in need of treatment" include mammals already having the disease or disorder, as well as those prone to having the disease or disorder, including those in which the disease or disorder is to be prevented.
[208] In one embodiment, an immunoglobulin disclosed herein is administered to a patient having a disease involving inappropriate expression of a protein or other molecule. Within the scope disclosed herein this is meant to include diseases and disorders characterized by aberrant proteins, due for example to alterations in the amount of a protein present, protein localization, posttranslational modification, conformational state, the presence of a mutant or pathogen protein, etc. Similarly, the disease or disorder may be characterized by alterations molecules including but not limited to polysaccharides and gangliosides. An overabundance may be due to any cause, including but not limited to overexpression at the molecular level, prolonged or accumulated appearance at the site of action, or increased activity of a protein relative to normal. Included within this definition are diseases and disorders characterized by a reduction of a protein. This reduction may be due to any cause, including but not limited to reduced expression at the molecular level, shortened or reduced appearance at the site of action, mutant forms of a protein, or decreased activity of a protein relative to normal. Such an overabundance or reduction of a protein can be measured relative to normal expression, appearance, or activity of a protein, and said measurement may play an important role in the development and/or clinical testing of the immunoglobulins disclosed herein.
[209] The immunoglobulins herein may be used to treat cancer. By "cancer" and
"cancerous" herein refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include but are not limited to carcinoma, lymphoma, blastoma, sarcoma (including liposarcoma),
neuroendocrine tumors, mesothelioma, schwanoma, meningioma, adenocarcinoma, melanoma, and leukemia or lymphoid malignancies.
[210] More particular examples of such cancers include hematologic malignancies, such as Hodgkin's lymphoma; non-Hodgkin's lymphomas (Burkitt's lymphoma, small lymphocytic lymphoma/chronic lymphocytic leukemia, mycosis fungoides, mantle cell lymphoma, follicular lymphoma, diffuse large B-cell lymphoma, marginal zone lymphoma, hairy cell leukemia and lymphoplasmacytic leukemia), tumors of lymphocyte precursor cells, including B-cell acute lymphoblastic leukemia/lymphoma, and T-cell acute lymphoblastic
leukemia/lymphoma, thymoma, tumors of the mature T and NK cells, including peripheral T- cell leukemias, adult T-cell leukemia/T-cell lymphomas and large granular lymphocytic leukemia, Langerhans cell histocytosis, myeloid neoplasias such as acute myelogenous leukemias, including AML with maturation, AML without differentiation, acute promyelocytic leukemia, acute myelomonocytic leukemia, and acute monocytic leukemias, myelodysplastic syndromes, and chronic myeloproliferative disorders, including chronic myelogenous leukemia; tumors of the central nervous system such as glioma, glioblastoma,
neuroblastoma, astrocytoma, medulloblastoma, ependymoma, and retinoblastoma; solid tumors of the head and neck (eg. nasopharyngeal cancer, salivary gland carcinoma, and esophagael cancer), lung (eg. small-cell lung cancer, non-small cell lung cancer,
adenocarcinoma of the lung and squamous carcinoma of the lung), digestive system (eg. gastric or stomach cancer including gastrointestinal cancer, cancer of the bile duct or biliary tract, colon cancer, rectal cancer, colorectal cancer, and anal carcinoma), reproductive system (eg. testicular, penile, or prostate cancer, uterine, vaginal, vulval, cervical, ovarian, and endometrial cancer), skin (eg. melanoma, basal cell carcinoma, squamous cell cancer, actinic keratosis), liver (eg. liver cancer, hepatic carcinoma, hepatocellular cancer, and hepatoma), bone (eg. osteoclastoma, and osteolytic bone cancers) additional tissues and organs (eg. pancreatic cancer, bladder cancer, kidney or renal cancer, thyroid cancer, breast cancer, cancer of the peritoneum, and Kaposi's sarcoma), and tumors of the vascular system (eg. angiosarcoma and hemagiopericytoma).
[211] The immunoglobulins herein may be used to treat autoimmune diseases. By
"autoimmune diseases" herein include allogenic islet graft rejection, alopecia areata, ankylosing spondylitis, antiphospholipid syndrome, autoimmune Addison's disease, antineutrophil cytoplasmic autoantibodies (ANCA), autoimmune diseases of the adrenal gland, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune myocarditis, autoimmune neutropenia, autoimmune oophoritis and orchitis, autoimmune
thrombocytopenia, autoimmune urticaria, Behcet's disease, bullous pemphigoid,
cardiomyopathy, Castleman's syndrome, celiac spruce-dermatitis, chronic fatigue immune disfunction syndrome, chronic inflammatory demyelinating polyneuropathy, Churg-Strauss syndrome, cicatrical pemphigoid, CREST syndrome, cold agglutinin disease, Crohn's disease, dermatomyositis, discoid lupus, essential mixed cryoglobulinemia, factor VIII deficiency, fibromyalgia-fibromyositis, glomerulonephritis, Grave's disease, Guillain-Barre, Goodpasture's syndrome, graft-versus-host disease (GVHD), Hashimoto's thyroiditis, hemophilia A, idiopathic pulmonary fibrosis, idiopathic thrombocytopenia purpura (ITP), IgA neuropathy, IgM polyneuropathies, immune mediated thrombocytopenia, juvenile arthritis, Kawasaki's disease, lichen plantus, lupus erthematosis, Meniere's disease, mixed connective tissue disease, multiple sclerosis, type 1 diabetes mellitus, myasthenia gravis, pemphigus vulgaris, pernicious anemia, polyarteritis nodosa, polychrondritis, polyglandular syndromes, polymyalgia rheumatica, polymyositis and dermatomyositis, primary
agammaglobinulinemia, primary biliary cirrhosis, psoriasis, psoriatic arthritis, Reynauld's phenomenon, Reiter's syndrome, rheumatoid arthritis, sarcoidosis, scleroderma, Sjorgen's syndrome, solid organ transplant rejection, stiff-man syndrome, systemic lupus
erythematosus, takayasu arteritis, temporal arteristis / giant cell arteritis, thrombotic thrombocytopenia purpura, ulcerative colitis, uveitis, vasculitides such as dermatitis herpetiformis vasculitis, vitiligo, and Wegner's granulomatosis.
[212] The immunoglobulins herein may be used to treat inflammtory disorders. By
"inflammatory disorders" herein include acute respiratory distress syndrome (ARDS), acute septic arthritis, adjuvant arthritis, juvenile idiopathic arthritis, allergic encephalomyelitis, allergic rhinitis, allergic vasculitis, allergy, asthma, atherosclerosis, chronic inflammation due to chronic bacterial or viral infectionis, chronic obstructive pulmonary disease (COPD), coronary artery disease, encephalitis, inflammatory bowel disease, inflammatory osteolysis, inflammation associated with acute and delayed hypersensitivity reactions, inflammation associated with tumors, peripheral nerve injury or demyelinating diseases, inflammation associated with tissue trauma such as burns and ischemia, inflammation due to meningitis, multiple organ injury syndrome, pulmonary fibrosis, sepsis and septic shock, Stevens- Johnson syndrome, undifferentiated arthropy, and undifferentiated spondyloarthropathy.
[213] Some autoimmune and inflammatory diseases that may be targeted by the immunoglobulins disclosed herein include Systemic Lupus Erythematosus, Rheumatoid arthritis, Sjogren's syndrome, Multiple sclerosis, Idiopathic thrombocytopenic purpura (ITP), Graves disease, Inflammatory bowel disease, Psoriasis, Type I diabetes, and Asthma.
[214] The immunoglobulins herein may be used to treat infectious diseases. By "infectious diseases" herein include diseases caused by pathogens such as viruses, bacteria, fungi, protozoa, and parasites. Infectious diseases may be caused by viruses including adenovirus, cytomegalovirus, dengue, Epstein-Barr, hanta, hepatitis A, hepatitis B, hepatitis C, herpes simplex type I, herpes simplex type II, human immunodeficiency virus, (HIV), human papilloma virus (HPV), influenza, measles, mumps, papova virus, polio, respiratory syncytial virus, rinderpest, rhinovirus, rotavirus, rubella, SARS virus, smallpox, viral meningitis, and the like. Infections diseases may also be caused by bacteria including Bacillus antracis, Borrelia burgdorferi, Campylobacter jejuni, Chlamydia trachomatis, Clostridium botulinum, Clostridium tetani, Diptheria, E. coli, Legionella, Helicobacter pylori, Mycobacterium rickettsia, Mycoplasma nesisseria, Pertussis, Pseudomonas aeruginosa, S. pneumonia, Streptococcus, Staphylococcus, Vibria cholerae, Yersinia pestis, and the like. Infectious diseases may also be caused by fungi such as Aspergillus fumigatus, Blastomyces dermatitidis, Candida albicans, Coccidioides immitis, Cryptococcus neoformans,
Histoplasma capsulatum, Penicillium marneffei, and the like. Infectious diseases may also be caused by protozoa and parasites such as chlamydia, kokzidioa, leishmania, malaria, rickettsia, trypanosoma, and the like.
[215] Furthermore, immunoglobulins disclosed herein may be used to prevent or treat additional conditions including but not limited to heart conditions such as congestive heart failure (CHF), myocarditis and other conditions of the myocardium; skin conditions such as rosecea, acne, and eczema; bone and tooth conditions such as bone loss, osteoporosis, Paget's disease, Langerhans' cell histiocytosis, periodontal disease, disuse osteopenia, osteomalacia, monostotic fibrous dysplasia, polyostotic fibrous dysplasia, bone metastasis, bone pain management, humoral malignant hypercalcemia, periodontal reconstruction, spinal cord injury, and bone fractures; metabolic conditions such as Gaucher's disease; endocrine conditions such as Cushing's syndrome; and neurological and neurodegenerative conditions such as Alzheimer's disease.
[216] Formulation
[217] Pharmaceutical compositions are contemplated wherein an immunoglobulin disclosed herein and one or more therapeutically active agents are formulated. Formulations of the immunoglobulins disclosed herein are prepared for storage by mixing said
immunoglobulin having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed., 1980, incorporated entirely by reference), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, acetate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride;
hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl orbenzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; sweeteners and other flavoring agents; fillers such as microcrystalline cellulose, lactose, corn and other starches; binding agents; additives; coloring agents; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or
polyethylene glycol (PEG). In one embodiment, the pharmaceutical composition that comprises the immunoglobulin disclosed herein may be in a water-soluble form, such as being present as pharmaceutically acceptable salts, which is meant to include both acid and base addition salts. "Pharmaceutically acceptable acid addition salt" refers to those salts that retain the biological effectiveness of the free bases and that are not biologically or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like. "Pharmaceutically acceptable base addition salts" include those derived from inorganic bases such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Some embodiments include at least one of the ammonium, potassium, sodium, calcium, and magnesium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. The formulations to be used for in vivo administration may be sterile. This is readily accomplished by filtration through sterile filtration membranes or other methods.
[218] The immunoglobulins disclosed herein may also be formulated as immunoliposomes. A liposome is a small vesicle comprising various types of lipids, phospholipids and/or surfactant that is useful for delivery of a therapeutic agent to a mammal. Liposomes containing the immunoglobulin are prepared by methods known in the art. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid
arrangement of biological membranes. Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising
phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
[219] The immunoglobulin and other therapeutically active agents may also be entrapped in microcapsules prepared by methods including but not limited to coacervation techniques, interfacial polymerization (for example using hydroxymethylcellulose or gelatin- microcapsules, or poly-(methylmethacylate) microcapsules), colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules), and macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed., 1980, incorporated entirely by reference. Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymer, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example poly(2- hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides, copolymers of L-glutamic acid and gamma ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the Lupron Depot® (which are injectable
microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), poly- D-(-)-3-hydroxybutyric acid, and ProLease® (commercially available from Alkermes), which is a microsphere-based delivery system composed of the desired bioactive molecule incorporated into a matrix of poly-DL-lactide-co-glycolide (PLG).
[220] Administration
[221] Administration of the pharmaceutical composition comprising an immunoglobulin disclosed herein, e.g., in the form of a sterile aqueous solution, may be done in a variety of ways, including, but not limited to orally, subcutaneously, intravenously, intranasally, intraotically, transdermally, topically (e.g., gels, salves, lotions, creams, etc.),
intraperitoneally, intramuscularly, intrapulmonary, vaginally, parenterally, rectally, or intraocularly. In some instances, for example for the treatment of wounds, inflammation, etc., the immunoglobulin may be directly applied as a solution or spray. As is known in the art, the pharmaceutical composition may be formulated accordingly depending upon the manner of introduction.
[222] Subcutaneous administration may be used in circumstances where the patient may self-administer the pharmaceutical composition. Many protein therapeutics are not sufficiently potent to allow for formulation of a therapeutically effective dose in the maximum acceptable volume for subcutaneous administration. This problem may be addressed in part by the use of protein formulations comprising arginine-HCI, histidine, and polysorbate.
Immunoglobulins disclosed herein may be more amenable to subcutaneous administration due to, for example, increased potency, improved serum half-life, or enhanced solubility. As is known in the art, protein therapeutics are often delivered by IV infusion or bolus. The immunoglobulins disclosed herein may also be delivered using such methods. For example, administration may be by intravenous infusion with 0.9% sodium chloride as an infusion vehicle.
[223] Pulmonary delivery may be accomplished using an inhaler or nebulizer and a formulation comprising an aerosolizing agent. For example, AERx® inhalable technology commercially available from Aradigm, or Inhance™ pulmonary delivery system commercially available from Nektar Therapeutics may be used. Furthermore, immunoglobulins disclosed herein may be amenable to oral delivery.
[224] In addition, any of a number of delivery systems are known in the art and may be used to administer the immunoglobulins disclosed herein. Examples include, but are not limited to, encapsulation in liposomes, microparticles, microspheres (e.g., PLA PGA microspheres), and the like. Alternatively, an implant of a porous, non-porous, or gelatinous material, including membranes or fibers, may be used. Sustained release systems may comprise a polymeric material or matrix such as polyesters, hydrogels,
poly(vinylalcohol),polylactides, copolymers of L-glutamic acid and ethyl-L-gutamate, ethylene-vinyl acetate, lactic acid-glycolic acid copolymers such as the Lupron Depot®, and poly-D-(-)-3-hydroxyburyric acid. It is also possible to administer a nucleic acid encoding an immunoglobulin disclosed herein, for example by retroviral infection, direct injection, or coating with lipids, cell surface receptors, or other transfection agents. In all cases, controlled release systems may be used to release the immunoglobulin at or close to the desired location of action.
[225] Dosing
[226] The dosing amounts and frequencies of administration are, in one embodiment, selected to be therapeutically or prophylactically effective. As is known in the art, adjustments for protein degradation, systemic versus localized delivery, and rate of new protease synthesis, as well as the age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition may be necessary, and will be ascertainable with routine experimentation by those skilled in the art.
[227] The concentration of the therapeutically active immunoglobulin in the formulation may vary from about 0.1 to 100 weight %. In one embodiment, the concentration of the immunoglobulin is in the range of 0.003 to 1 .0 molar. In order to treat a patient, a
therapeutically effective dose of the immunoglobulin disclosed herein may be administered. By "therapeutically effective dose" herein is meant a dose that produces the effects for which it is administered. The exact dose will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques. Dosages may range from 0.0001 to 100 mg/kg of body weight or greater, for example 0.1 , 1 , 10, or 50 mg/kg of body weight. In one embodiment, dosages range from 1 to 10mg/kg.
[228] In some embodiments, only a single dose of the immunoglobulin is used. In other embodiments, multiple doses of the immunoglobulin are administered. The elapsed time between administrations may be less than 1 hour, about 1 hour, about 1-2 hours, about 2-3 hours, about 3-4 hours, about 6 hours, about 12 hours, about 24 hours, about 48 hours, about 2-4 days, about 4-6 days, about 1 week, about 2 weeks, or more than 2 weeks.
[229] In other embodiments the immunoglobulins disclosed herein are administered in metronomic dosing regimes, either by continuous infusion or frequent administration without extended rest periods. Such metronomic administration may involve dosing at constant intervals without rest periods. Typically such regimens encompass chronic low-dose or continuous infusion for an extended period of time, for example 1 -2 days, 1-2 weeks, 1-2 months, or up to 6 months or more. The use of lower doses may minimize side effects and the need for rest periods. [230] In certain embodiments the immunoglobulins disclosed herein and one or more other prophylactic or therapeutic agents are cyclically administered to the patient. Cycling therapy involves administration of a first agent at one time, a second agent at a second time, optionally additional agents at additional times, optionally a rest period, and then repeating this sequence of administration one or more times. The number of cycles is typically from 2 - 10. Cycling therapy may reduce the development of resistance to one or more agents, may minimize side effects, or may improve treatment efficacy.
[231] Combination Therapies
[232] The immunoglobulins disclosed herein may be administered concomitantly with one or more other therapeutic regimens or agents. The additional therapeutic regimes or agents may be used to improve the efficacy or safety of the immunoglobulin. Also, the additional therapeutic regimes or agents may be used to treat the same disease or a comorbidity rather than to alter the action of the immunoglobulin. For example, an immunoglobulin disclosed herein may be administered to the patient along with chemotherapy, radiation therapy, or both chemotherapy and radiation therapy.
[233] The terms "in combination with" and "co-administration" are not limited to the administration of said prophylactic or therapeutic agents at exactly the same time. Instead, it is meant that the immunoglobulin disclosed herein and the other agent or agents are administered in a sequence and within a time interval such that they may act together to provide a benefit that is increased versus treatment with only either the immunoglobulin disclosed herein or the other agent or agents. In some embodiments, immunoglobulins disclosed herein and the other agent or agents act additively, and sometimes
synergistically. Such molecules are suitably present in combination in amounts that are effective for the purpose intended. The skilled medical practitioner can determine empirically, or by considering the pharmacokinetics and modes of action of the agents, the appropriate dose or doses of each therapeutic agent, as well as the appropriate timings and methods of administration.
[234] The immunoglobulin disclosed herein may be administered in combination with one or more other prophylactic or therapeutic agents, including but not limited to cytotoxic agents, chemotherapeutic agents, antibiotics, antifungal agents, antiviral agents, cytokines, growth inhibitory agents, anti-hormonal agents, kinase inhibitors, anti-angiogenic agents, cardioprotectants, immunostimulatory agents, immunosuppressive agents, agents that promote proliferation of hematological cells, angiogenesis inhibitors, protein tyrosine kinase (PTK) inhibitors, other antibodies, Fc fusions, or immunoglobulins, or other therapeutic agents. The therapies of the invention may be combined with other immunotherapies. The therapies of the invention may be combined with antagonists of chemokines or cytokines, including but not limited to antibodies and Fc fusions.
[235] The immunoglobulins disclosed herein may be combined with other therapeutic regimens. For example, in one embodiment, the patient to be treated with an
immunoglobulin disclosed herein may also receive radiation therapy. Radiation therapy can be administered according to protocols commonly employed in the art and known to the skilled artisan. Such therapy includes but is not limited to cesium, iridium, iodine, or cobalt radiation. The radiation therapy may be whole body irradiation, or may be directed locally to a specific site or tissue in or on the body, such as the lung, bladder, or prostate. Optionally, the radiation therapy may be administered as a single dose or as multiple, sequential doses. The skilled medical practitioner can determine empirically the appropriate dose or doses of radiation therapy useful herein. In accordance with another, an immunoglobulin disclosed herein and one or more other anti-cancer therapies are employed to treat cancer cells ex vivo. It is contemplated that such ex vivo treatment may be useful in bone marrow transplantation and particularly, autologous bone marrow transplantation. For instance, treatment of cells or tissue(s) containing cancer cells with immunoglobulin and one or more other anti-cancer therapies, such as described above, can be employed to deplete or substantially deplete the cancer cells prior to transplantation in a recipient patient. It is of course contemplated that the immunoglobulins disclosed herein may employ in combination with still other therapeutic techniques such as surgery.
[236] All cited references are herein expressly incorporated by reference in their entirety.
[237] Whereas particular embodiments of the invention have been described above for purposes of illustration, it will be appreciated by those skilled in the art that numerous variations of the details may be made without departing from the invention as described in the appended claims.
EXAMPLES
[238] Examples are provided below to illustrate the present invention. These examples are not meant to constrain the present invention to any particular application or theory of operation.
[239]
[240] Example 1 . Design of Heterodimeric Fc Regions
[241] Variants that promote Fc heterodimerization and disfavor homodimerization were designed using a combination of computational strutu re-based and rational protein engineering approaches. Computational structure-based methods (PDA® technology) were used to evaluate possible amino acid substitutions in the CH3 region for their ability to stabilize Fc heterodimers and destabilize Fc homodimers . Amino acid energies for selected pairs of positions were calculated in the context of the heterodimer (both variant amino acids) and the two homodimers (variant amino acid at one position and the wild-type amino acid at the other). Energies were normalized against the calculated energy of the wild-type amino acid pair. The preference for the heterodimeric species was calculated using the following two equations:
[242] Heterodimer Preference 1 =
E(heterodimer) - max [ E(homodimer 1 ), E(homodimer 2) ]
[243] Heterodimer Preference 2 =
E(heterodimer) - min [ E(homodimer 1 ), E(homodimer 2) ]
[244] Of particular interest are those variants for which one or both of the heterodimer preferences are negative. This indicates that the heterodimer is predicted to be more stable than one or both of the homodimers.
[245] Example 2. Screening of Variant Fc Regions for Fc Heterodimerization
[246] In order to screen for Fc variants that favor heterodimerization, an empty-Fc/scFv-Fc system was developed as a test system, illustrated in Figure 1. DNA constructs were designed encoding two different immunoglobulin polypeptides: (i) scFv-Fc (VH-linker-VK- Hinge-CH2-CH3') and (ii) empty-Fc (Hinge-CH2-CH3"). Here the two different CH3 domains, CH3' (prime) and CH3" (double prime), represent the different variant versions that were designed to promote heterodimer formation and discourage homodimer formation. As shown in Figure 1 , cotransfection and expression of these two polypeptides results in three possible dimers. Each of the three dimers is composed of a unique number of immunoglobulin (Ig) domains and thus unique molecular weights. Consequently, the population of each species can be visualized using gel electrophoresis. Figure 2 provides amino acid sequences of the scFv-Fc and empty-Fc constructs, along with sequences of native IgG's.
[247] The scFv-Fc/empty-Fc system was tested using wild-type lgG1 Fc regions and an scFv derived from a murine anti-CD3 antibody OKT3 (Kjer-Nielsen et al., 2004, Proc Natl Acad Sci USA 101 :7675-7680). DNA encoding the OKT3 scFv was generated by gene sythesis (Integrated DNA Technologies). All constructs were subcloned into the expression vector pTT5 (Durocher et al., 2002, Nucleic acids research 30: E9) using standard molecular biology techniques. scFv-Fc and empty-Fc constructs with native lgG1 Fc regions were cotransfected into HEK293E cells (Durocher et al., 2002, Nucleic Acids Research 30: E9) for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography. The protein species generated were visualized electrophoretically using an Agilent 2100 Bioanalyzer microfluidics-based platform (Agilent Technologies) . Agilent chips for SDS-PAGE were run on the BioAnalyzer using kit# 5067-1517, lot# NA06BP01 containing gels, chips, and buffers, carried out using the manufacturer's instructions.
Samples were run in duplicate, one under reducing and the other under non-reducing conditions. Results are shown in Figure 3. The 240 kilodalton (KDa) band in all of the gel lanes represents a protein in the loading buffer used as a normalization standard. Under reducing conditions (lanes 1-5), two bands were observed, corresponding to scFv-Fc monomer and empty-Fc monomer. Under non-reducing conditions (lanes 6-10), three bands were observed, corresponding to scFv-Fc homodimer, scFc-Fv/empty-Fc heterodimer, and empty-Fc homodimer. This result confirmed the ability to detect each of the different protein species, indicating the scFv-Fc/empty-Fc system provided a good system for quantification of the degree of heterodimerization from designed Fc variants.
[248] A DNA library encoding variant immunoglobulin polypeptides, designed as described above, was constructed in the scFv-Fc and empty-Fc constructs. Pairs of scFv-Fc and empty-Fc constructs were cotransfected into HEK293E cells for expression, and resulting proteins were purified using protein A affinity chromatography. Relative concentrations of the expressed protein species were determined using the Agilent 2100 Bioanalyzer
microfluidics-based platform, as described above. Resulting gels are shown in Figure 4. A quantitative summary of the results from this library is provided in Figure 5. A number of designed variants increased the content of heterodimer relative to that of the native parent Fc region.
[249] Using the information obtained from the previous library, a new library was designed and screened as described above. Quantitative electrophoretic results are shown in Figures 6 and 7. A number of designed variants increased the content of heterodimer relative to that of the native parent Fc region. Preferred and most preferred variants from this screen are listed in Tables 1 and 2 respectively.
Table 1. Preferred CH3 domain variants that favor Fc heterodimerization.
Variant 1 Variant 2
F405A T394F
S364D Y349K
S364E L368K
S364E Y349K
S364F K370G
S364H Y349K S364H Y349T
S364Y K370G
T411K K370E
V397S/F405A T394F
K370R/T411K K370E/T411E
L351E/S364D Y349K/L351K
L351E/S364E Y349K/L351K
L351E/T366D L351K/T366K
P395T/V397S/F405A T394F
S364D/K370G S364Y/K370R
S364D/T394F Y349K/F405A
S364E/F405A Y349K/T394F
S364E/F405S Y349K/T394Y
S364E/T411E Y349K/D401K
S364H/D401K Y349T/T411E
S364H/F405A Y349T/T394F
S364H/T394F Y349T/F405A
Y349C/S364E Y349K/S354C
L351E/S364D/F405A Y349K/L351K/T394F
L351K/S364H/D401K Y349T/L351E/T411E
S364E/T411E/F405A Y349K/T394F/D401K
S364H/D401K/F405A Y349T/T394F/T411E
S364H/F405A/T411E Y349T/T394F/D401K
Table 2. Most preferred CH3 domain variants that favor Fc heterodimerization.
Variant 1 Variant 2
F405A T394F
S364D Y349K
S364E Y349K
S364H Y349T
L351K L351E
D401K T411E
S364D/T394F Y349K/F405A
S364E/F405A Y349K/T394F
S364H/D401K Y349T/T411E
S364H/F405A Y349T/T394F
S364H/T394F Y349T/F405A
L351K/S364H/D401K Y349T/L351E/T411E
S364H/D401K/F405A Y349T/T394F/T411E
S364H/F405A/T411E Y349T/T394F/D401K
[250] Example 3. Description of mAb-Fv and mAb-Fab immunoglobulin formats
[251] Heterodimerization of distinct immunoglobulin heavy chains provides a novel beneficial approach to bispecific co-targeting of antigens. The capacity to co-engage multiple antigens, particularly the capacity to link binding of a target antigen to activation of a cellular receptor, offers the potential to enhance the clinical efficacy of antibody therapeutics, as well as engineer novel mechanisms of action. Yet an important property of bispecifics that engage many immune receptors is activation only in proximity to the primary or first target antigen. Biologically, the immune system accomplishes this naturally via monovalent binding of an immune receptor to its cognate ligand, often at low affinity. Cellular activation occurs only by cross-linking, achieved typically by antibody/antigen immune complexes, or via effector cell to target cell engagement. For some immune receptors, for example the CD3 signaling receptor on T cells, activation only upon engagement with co-engaged target is critical, as nonspecific cross-linking can elicit a cytokine storm and toxicity (Perruche et al., 2009, J Immunol 183[2]:953-61 ). An engineering solution, as described here, is to engage such immune receptors monovalently rather than multivalently such that activation occurs in response to cross-linking only in the presence of the primary target antigen.
[252] Example 4. mAb-Fv and mAb-Fab immunoglobulins that monovalently co-engage FcyRllla
[253] The low affinity Fc gamma receptors (FcyRs) such as FcyRlla, FcyRllb, and FcyRllla bind monovalently to the antibody Fc region. Monovalent binding does not activate cells expressing these FcyRs; however, cell-to-cell contact leads to receptor cross-linking and clustering on the cell surface, leading to activation. For FcyRllla (CD16) expressed on natural killer (NK) cells, target cell killing is mediated by receptor cross-linking and cellular activation, which occurs when the effector cell engages the target cell in a highly avid format (Bowles & Weiner, 2005, J Immunol Methods 304:88-99, Marquez et al., 2010, Cell Immunol 264[1 ]:86-92; expressly incorporated by reference). In this regard, FcyRllla co-targeting is a particularly good application of the mAb-Fv format, which is capable of engaging target antigen bivalently while co-engaging FcyRllla monovalently.
[254] mAb-Fv and mAb-Fab immunoglobulins that co-target FcyRllla were constructed using the variable region of the anti- human FcyRllla antibody 3G8 as Fv-2. A variety of constructs were generated that target various primary antigens (antigen-1 ), including the solid tumor antigens Her2 and EGFR, and the myeloma antigen HM1 .24. Antibody variable regions targeting these antigens were obtained from the anti-Her2 antibody trastuzumab (Carter et al., 1992, Proc Natl Acad Sci USA 89:4285-4289), a humanized version of the anti-EGFR antibody C225 (USSN1 1/004,054), and a humanized version of the anti-HM1.24 antibody HM1 .24 (USSN1 1/857,310). The different constructs incorporated a variety of linker compositions and lengths, and a number of them incorporated hetero-Fc variants that favor heterodimerization. In addition, in order to engineer selectivity to FcyRllla relative to other Fc receptors, several constructs utilized knockout Fc variants that ablate binding to FcyRs and complement. In addition, some constructs incorporated detection/purification tags on the C- termini, including His-tags (HHHHHH) and Flag-tags (DYKDDDDK). The anti-FcyRllla (otFcyRllla) mAb-Fv and mAb-Fab constructs are listed in Table 3 along with a description of their relevant components. Each immunoglobulin is given an identification number (ID). Amino acid sequences of these immunoglobulins are listed in Figure 9. Also included in Table 3 are control antibodies used as controls and comparators in the anti-Her2 experiments. These include an lgG1 version of trastuzumab, as well as variants with ablated (G236R/L328R) and enhanced (S239D/I332E) binding to FcyR's, including FcyRllla.
Table 3. mAb-Fv and mAb-Fab constructs and control antibodies for FcyRllla co-targeting
ID Ag-1 x Ag-2 Fv-1a FcR Binding13 Hetero-Fcc Linkerd Fv-2
9064 HER2 x FcyRII la trastuzumab Native lgG1 Homo Linker 1 3G8
9074 HER2 x FcyRII la trastuzumab Native lgG1 Homo Linker 2 3G8
9375 HER2 x FcyRII la trastuzumab Native lgG1 Hetero TF-HA Linker 2 3G8
9387 EGFR x FcyRII la humanized 225 Native lgG1 Hetero TF-HA Linker 2 3G8
9516 HER2 x FcyRII la trastuzumab G236R/L328R Hetero TF-HA Linker 2 3G8
9517 EGFR x FcyRII la hu 225 G236R/L328R Hetero TF-HA Linker 2 3G8
9518 HM1 .24 X FcyRllla hu aHM1.24 G236R/L328R Hetero TF-HA Linker 2 3G8
8427 HER2 only trastuzumab Native lgG1 Homo None None
8428 HER2 only trastuzumab S239D/I332E Homo None None
8429 HER2 only trastuzumab G236R/L328R Homo None None
4614 RSV motavizumab S239D/I332E Homo None None a hu = humanized
b G236R/L328R is an Fc knockout variant that ablates binding to all FcyRs and complement, and ablates FcyR- and complement- mediated effector function. S239D/I332E enhances affinity to activating FcyRs, and improves FcyR- mediated effector function.
c Homo = Homodimeric native IgG Fc region. Hetero TF-HA = Y349T/T394F and
S364H/F405A variant CH3 domains that favor heterodimerization and disfavor
homodimerization.
d Linker 1 = SSDKTHTSPPSPGGGGSGGGG (SEQ ID NO. 43)
Linker 2 = SSDKTHTSPPSPGGGGSGGGGSGGGGSGGGG (SEQ ID NO. 44)
[255] The immunoglobulins in Table 3 were constructed using a combination of gene synthesis (Blue Heron Biotechnology, Bothell, WA), PCR subcloning, quick-change mutagenesis, and other standard molecular biology methods. All DNA was constructed in the context of the expression vector pTT5 (Durocher et al., 2002, Nucleic acids research 30: E9) using standard molecular biology techniques. Heavy and light chains for each
immunoglobulin were cotransfected into HEK293E cells (Durocher et al., 2002, Nucleic Acids Research 30: E9) for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography.
[256] The proteins were visualized electrophoretically using the Agilent 2100 Bioanalyzer microfluidics-based platform (Agilent Technologies). Agilent chips for SDS-PAGE were run on the BioAnalyzer using kit# 5067-1517, lot# NA06BP01 containing gels, chips, and buffers, carried out using the manufacturer's instructions. Samples were in duplicate, one under reducing and the other under non-reducing conditions. Representative gels are shown in Figure 10. As expected, under reducing conditions three bands were observed at the approximate calculated molecular weights, corresponding to the light chain (-25 KDa) and two heavy chains (-65 KDa) for each mAb-Fv. Under non-reducing conditions a single main band at -180 KDa was observed for each mAb-Fv and -200 KDa for the mAb-Fab, corresponding to the tetrameric immunoglobulin (two heavy chains, each complexed with a light chain). The results are consistent for the three anti-FcyRllla constructs co-targeting Her2, EGFR, and HM1.24, indicating the broad applicability of the mAb-Fv format for co- engaging targets.
[257] The immunoglobulins were analyzed using size-exclusion chromatography (SEC) to measure their size (more correctly, their hydrodynamic volume) and determine the nativelike behavior of the purified samples. These experiments used an Agilent 1200 High- performance liquid chromatography (HPLC) system. Samples were injected onto a
Superdex™ 200 10/300 GL column (model# 17-5175-01 , lot# 10034171 ) (Amersham) at 0.5ml_s/min using 1X PBS, pH 7.4 as the mobile phase. Results for select mAb-Fv samples are shown in Figure 1 1. A molecular weight standard was run (not shown). All of the mAb-Fv samples showed a main peak at an elution time that, based on the molecular weight standard, corresponded to the approximate molecular weight of the tetrameric mAb-Fv heterodimer heavy chain species (-180 KDa). The symmetry of the peaks and the relatively low populations of other species for all of the anti-FcyRllla mAb-Fv's (aHer2, aEGFR, and aHM1.24) indicated that the immunoglobulin format is robust.
[258] The 9516 anti-Her2 x anti-FcyRllla (aHer2 x aFcyRllla) mAb-Fv was constructed in the context of an IgGI comprising the knockout Fc variant (Fc KO) G236R/L328R in order to engineer selective FcyRllla binding. In other words, binding of the Fc region to all of the FcyRs and complement was ablated, and then only FcyRllla binding was built back as antigen-2 in the mAb-Fv format. Binding of 9516 and control antibodies (Native IgGI and Fc KO) to the relevant human FcyR's was tested using surface plasmon resonance (Biacore®). Binding measurements were performed using a Biacore® 3000 instrument (Biacore®). Antibodies were captured onto an immobilized Protein A (Pierce) CM5 biosensor chip (Biacore®), generated using a standard primary amine coupling protocol. All measurements were performed in HBS-EP (0.01 M HEPES pH 7.4, 0.15 M NaCI, 3 mM EDTA, 0.005% v/v surfactant P20), and glycine buffer (10 mM glycine-HCI, pH 1.5) was used for the Protein A surface regeneration. All antibodies (50 nM in HBS-EP) were immobilized on the Protein A surface. Fc receptors were injected over antibody bound surface, followed by a dissociation phase. After each cycle, the Protein A surface was regenerated by injecting glycine buffer (pH 1.5). Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals (response of reference channel and injection of running buffer). The results in Figure 12 show that while native lgG1 binds to all of the FcyRs, the Fc KO variant is ablated for binding to all of the FcyRs. Similar to the Fc KO, the 9516 aHer2 x aFcyRNIa mAb-Fv is ablated for binding to FcyRI, FcyRlla, and FcyRI lb; however, in contrast to the knockout variant antibody, 9516 has strong affinity for FcyRNIa, due to the engineered Fv-2 binding site. The kinetic off-rate of dissociation (koff) for FcyRNIa binding is slower than that of native lgG1 , a consequence of the greater affinity of the 3G8 variable region for the receptor relative to the lgG1 Fc region.
[259] The neonatal Fc receptor (FcRn) protects IgG from degradation and is responsible in part for the long half-life (-21 days for lgG1 ) of antibodies in circulation. FcRn binds at a site on the Fc region separate from the binding site for FcyRs, and the Fc KO variant
G236R/L328R does not impact FcRn affinity. Binding to FcRn by the 9516 mAb-Fv and control antibodies was tested using a Biacore method similar to above, except that a Her2 antigen capture chip format was used (the protein A binding site on Fc overlaps with the site for FcRn). As can be seen in Figure 13, binding of the mAb-Fv to FcRn was uncompromised relative to native lgG1 and the Fc KO variant.
[260] Affinity of the mAb-Fv for FcyRNIa was measured using Biacore®. The protein A capture format was used as described above, except that sensorgrams were obtained for a concentration series of receptor analtye in order to calculate an accurate affinity. Figure 14 shows Biacore® sensorgrams depicting the binding of various concentrations of the V158 isoform of FcyRNIa to protein A-captured 9516 mAb-Fv. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate
nonspecific signals (response of reference channel and injection of running buffer). The equilbrium constant was calculated by global fitting of binding data with a 1 :1 Langmuir binding model using BIAevaluation software. The aHer2 x aFcyRNIa mAb-Fv bound FcyRNIa with an equilibrium constant of dissociation (Kd) of 1 10 nM, approximately 2.5-fold tighter than the 280 nM affinity of native lgG1 (Richards et al., 2008, Mol Cancer Ther 7[8]:2517- 27), consistent with the slower dissociation observed relative to native lgG1 (Figure 12).
[261] Co-engagement of Her2 and FcyRNIa by the 9516 mAb-Fv was confirmed using a Biacore experiment similar to as described above, but using Her2 capture instead of protein A. 100 nM concentration of the 9516 mAb-Fv was captured to a HER2 chip, followed by injection of 200 nM V158 FcyRNIa analyte. Data were processed by zeroing time and response before the injection of 9516. The results in Figure 15 demonstrate the
simultaneous co-engagement of Her2 by the Fv-1 region and FcyRNIa by the Fv-2 region. Engagement of Fc receptor is not due to binding by the Fc region, which due to the Fc KO variant is ablated for binding to all of the Fc receptors (Figure 12).
[262] Similar binding experiments were carried out for the aHM1 .24 x aFcyRl lla mAb-Fv. The protein A capture format was used with a receptor analyte concentration series to calculate the affinity of the 9518 mAb-Fv for V158 FcyRl l la. Figure 16 shows Biacore® sensorgrams depicting the binding of receptor at various concentrations to protein A- captured 9518 mAb-Fv. Data were processed by zeroing time and response before the injection of receptor and by subtracting appropriate nonspecific signals. The equilbrium constant was calculated by global fitting of binding data with a 1 :1 Langmuir binding model using BIAevaluation software. The aHM1 .24 x aFcyRl lla mAb-Fv bound V158 FcyRl l la with an equilibrium constant of dissociation (Kd) of 96 nM, approximately 2.9-fold tighter than the 280 nM affinity of native lgG1 (Richards et al., 2008, Mol Cancer Ther 7[8]:2517-27).
[263] Co-engagement of HM1 .24 and FcyRl l la by the 9518 mAb-Fv was confirmed using the antigen capture Biacore® method as described above. 100 nM concentration of the 9518 mAb-Fv was captured to a HM1 .24 chip, followed by injection of 200 nM V158 FcyRll la analyte. Data were processed by zeroing time and response before the injection of 9516. The results in Figure 17 demonstrate the simultaneous co-engagement of HM1 .24 by the Fv- 1 region and FcyRl l la by the Fv-2 region.
[264] A cytotoxicity assay was carried out to test the capacity of the aHer2 x aFcyRl lla mAb-Fv to mediate cellular killing against Her2-expressing cells. A native lgG1 version of the anti-Her2 antibody trastuzumab was run as a control, and an Fc variant version
(S239D/I332E) with enhanced FcyRl l la binding was also tested as a comparator. An anti- respiratory syncytial virus (RSV) antibody with the variable region of motavizumab, with the S239D/I332E Fc region served as a negative control for lack of binding to Her2. Human peripheral blood mononuclear cells (PBMC) were purified from leukopacks using a Ficoll™ gradient. Her2+ SK-BR-3 breast carcinoma cells were seeded into 96-well plates and opsonized with mAb-Fv or control/comparator antibodies antibodies at the indicated concentrations. Triton X-100 and PBMC alone were run as controls. Effector cells were added at 50: 1 PBMC/target cells, and plates were incubated at 37 °C for 4 h. Cell lysis was measured by lactate dehydrogenase release using the Cytotox-ONE™ Homogeneous Membrane Integrity Assay (Promega). Cells were incubated with lactate dehydrogenase reaction mixture for 10 min., and fluorescence was measured using a Wallac Victor2 fluorometer (Perkin-Elmer). Fluorescence was normalized to minimal (PBMC alone) and maximal (Triton) lysis, and fit to a sigmoidal dose-response. The results are shown in Figure 18. The aHer2 x aFcyRll la mAb-Fv mediated a level of lysis substantially enhanced relative to native lgG1 trastuzumab, and comparable to the Fc-enhanced variant S239D/I332E. The results confirm the high affinity FcyRllla (antigen-2) binding mediated by novel engineered Fv-2 binding site, and demonstrate the capacity of the mAb-Fv format for enabling selective and optimal functions.
[265] Example 5. mAb-Fv immunoglobulins that monovalently co-engage FcyRllb
[266] The inhibitory receptor FcyRllb downregulates cellular activation of B cells, monocytes, mast cells, and other immune cell types. On B cells, inhibitory activity is elicited only when FcyRllb rengages into an immune complex with the cell surface B-cell receptor (BCR), a mechanism that is mediated by immune complexation of soluble IgG's with the same antigen that is recognized by the BCR (Heyman 2003, Immunol Lett 88[2]:157-161 ; Smith and Clatworthy, 2010, Nature Reviews Immunology 10:328-343; expressly
incorporated by reference). We explored the applicability of the mAb-Fv format for engaging FcyRllb monovalently while bivalently co-engaging an antigen in the BCR complex, specifically CD19. This immunomodulatory co-engagement mechanism is described in USSN 12/156, 183, filed May 30, 2008, entitled "Methods and Compositions for Inhibiting CD32b Expressing Cells", herein expressly incorporated by reference. Other co-targets for the FcyRllb-mediated immunomodulatory mechanism include other antigens involved with the BCR or components of its coreceptor complex, including for example CD21 , CD22, CD72, CD79a, CD79b, and CD81 , as described in USSN 12/156, 183.
[267] mAb-Fv immunoglobulins that co-target FcyRllb were constructed using as Fv-2 the variable region of 2B6, an antibody selective for the inhibitory receptor FcyRllb as Fv-2 (Veri et al., 2007, Immunology 121 :392-404). Targeting of CD19 antigen by Fv-1 utilized a humanized affinity matured version of the anti-CD19 antibody 4G7 described previously (USSN 12/156,183). The different constructs incorporated a gly-ser repeat linker, and knockout Fc variants in order to engineer selectivity to FcyRllb relative to other Fc receptors. The mAb-Fv constructs utilized either native CH3 domains (ID 9506) or hetero-Fc variants ID 9547). The anti-FcyRllb (otFcyRllb) mAb-Fv and constructs are listed in Table 4 along with a description of their relevant components. Amino acid sequences of these immunoglobulins are listed in Figure 19. Also included in Table 4 is a control antibody (ID 6187) containing the knockout variant used in these experiments.
Table 4. mAb-Fv constructs and control antibodies for FcyRllb co-targeting
ID Ag-1 x Ag-2 Fv-1a FcR Binding13 Hetero-Fcc Linkerd Fv-2
9506 CD19 x FcyRllb hu 4G7 G236R/L328R Homo Linker 3 2B6
9547 CD19 x FcyRllb hu 4G7 G236R/L328R Hetero TF-HA Linker 3 2B6
6187 CD19 only hu 4G7 G236R/L328R Homo None None a hu = humanized
b G236R/L328R is an Fc knockout variant that ablates binding to all FcyRs and complement, and ablates FcyR- and complement- mediated effector function.
0 Homo = Homodimeric native IgG Fc region. Hetero TF-HA = Y349T/T394F and
S364H/F405A variant CH3 domains that favor heterodimerization and disfavor
homodimerization.
d Linker 3 = GGGGSGGGGSGGGGSGGGGSGGGGSGGGG (SEQ ID NO. 45)
[268] The immunoglobulins in Table 4 were constructed as described above in the pTT5 expression vector. Heavy and light chains for each immunoglobulin were cotransfected into HEK293E cells for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography, as described above.
[269] The proteins were analyzed using electrophoresis using the Agilent Bioanalyzer as described above. SDS-PAGE results are shown in Figure 20. Although the 9506 construct comprising the homodimeric Fc region expressed poorly, faint bands could nonetheless be observed approximately at the expected molecular weights of the light chain (-25 KDa) and two heavy chains (-65 KDa). Likewise, under non-reducing conditions a single main band was observed for at the expected molecular weight of the tetrameric protein (two heavy chains, each complexed with a light chain, -180 KDa). Proper assembly of the mAb-Fv was supported by the SEC results (data not shown). The 9547 construct was identical 9506 except that it contained the Fc variants Y349T/T394F on one heavy chain and S364H/F405A on the other heavy chain to favor heterodimerization. Multiple bands were observed for 9547 in the SDS-PAGE gels. Bands were observed approximately at the expected molecular weights of the light chain and two heavy chains, and under non-reducing conditions a band was observed at the expected molecular weight of the tetrameric immunoglobulin. Assembly of the mAb-Fv was supported by the presence of an SEC peak that corresponded to the approximate molecular weight of the tetrameric mAb-Fv heterodimer species (data not shown). The size of the additional band in the non-reducing gel suggested a significant population of a monomeric heavy chain / light chain pair (-90 KDa). Thus due to the hetero- Fc variants, heavy chain monomers (complexed with light chain) are observed rather than homodimeric heavy chain species. Based on the intensity of the bands in the gel, this result appears to be due to differential expression between the two heavy chains. We suspect the poor expression of one of the heavy chains relative to the other is due to a glycosylation site in the 2B6 VL region (USSN12/349,876). Regardless, the results from the gels and SEC indicate a significant population of the designed heterodimeric mAb-Fv species. [270] Affinity of the 9547 aCD19 x aFcyRllb mAb-Fv was measured using Biacore® as described above. Figure 21 shows Biacore sensorgrams depicting the binding of various concentrations of FcyRllb to protein A-captured 9547 mAb-Fv. Data were processed and fitted as described above. The aCD19 x aFcyRllb mAb-Fv bound FcyRllb with a Kd of 44 nM, approximately 52-fold tighter than the 2.3 μΜ affinity of native lgG1 (Richards et al., 2008, Mol Cancer Ther 7[8]:2517-27). Because 9547 was constructed in the context of the knockout Fc variant G236R/L328R, binding to of the mAb-Fv is selective for FcyRllb, i.e. it binds the inhibitory receptor FcyRllb with high affinity but does not bind any of the activating FcyRs.
[271] The 9547 mAb-Fv and 6187 control antibody were assayed for capacity to inhibit BCR-mediated B cell activation as a result of BCR/FcyRllb co-engagement. This mechanism of B cell inhibition has been described previously (USSN 12/156, 183). Intracellular calcium mobilization was used as a quantitative measure of BCR-mediated B cell activation.
Intracellular free calcium concentration ([Ca2+]) was measured by flow cytometry using a Fluo-4 NW calcium assay (Molecular Probes, Eugene, OR). PBMCs were resuspended in calcium assay buffer and pre-loaded with Fluo-4 dye for 30 min at room temperature. After incubation with immunoglobulin of interest, cells were stimulated by addition of 10 μg/ml of anti-CD79b antibody. Calcium flux kinetics was recorded using a FACSCanto II flow cytometer and data were analyzed using FlowJo software (Tree Star, Ashland, OR). The data are plotted as the change of Mean Fluo-4 Intensity (MFI) over time. The results are shown in Figure 22. 9547 aCD19 x aFcyRllb mAb-Fv inhibited calcium mobilization induced by BCR crosslinking relative to the 6187 Fc KO aCD19 antibody. This activity is attributed to the high affinity (44 nM) FcyRllb co-engagement of the inhibitory receptor with BCR- associated CD19 antigen. The result further demonstrates the capacity of the mAb-Fv format for enabling selective and novel functional activities.
[272] Example 6. mAb-Fv immunoglobulins that monovalently co-engage CD3
[273] CD3 activation of T-cells occurs only when its associated T-cell receptor (TCR) engages antigen-loaded MHC on antigen presenting cells in a highly avid cell-to-cell synapse (Kuhns et al., 2006, Immunity 24:133-139). Because cross-linking of CD3 using an anti-CD3 antibody elicits a cytokine storm and toxicity (Perruche et al., 2009, J Immunol 183[2]:953-61 ; Chatenoud & Bluestone, 2007, Nature Reviews Immunology 7:622-632; expressly incorporated by reference), for practical clinical use, the preferred mode of CD3 co-engagement for redirected killing of targets cells is monovalent binding that results in activation only upon engagement with the co-engaged primary target. The mAb-Fv format is particularly well-suited for this mechanistic appproach. We explored the applicability of the mAb-Fv format for engaging CD3 monovalently while bivalently co-engaging tumor cell antigens.
[274] mAb-Fv immunoglobulins that co-target CD3 were constructed using as the Fv-2 variable region a humanized version of the anti-CD3 antibody OKT3. Fv-1 regions targeted the antigens Her2, HM1 .24, and CD19 using the variable regions described above. A variety of constructs were generated co-targeting these primary antigens bivalently and CD3 monovalently that incorporated different linker compositions and lengths, hetero-Fc variants that favor heterodimerization, and knockout Fc variants to ablate binding to FcyRs and complement. Constructs also incorporated His- and Flag- tags at the C-termini as potential detection/purification tags. The aCD3 mAb-Fv constructs are listed in Table 5 along with a description of their relevant components. Amino acid sequences of these immunoglobulins are listed in Figure 23.
Table 5. mAb-Fv constructs and control antibodies for CD3 co-targeting
ID Ag-1 x Ag-2 Fv-1a FcR Binding13 Hetero-Fcc Linkerd Fv-2a
9344 HER2 x CD3 trastuzumab Native lgG1 Homo Linker 4 hu OKT3
9376 HER2 x CD3 trastuzumab Native lgG1 Hetero TF-HA Linker 4 hu OKT3
9509 HER2 x CD3 trastuzumab G236R/L328R Homo Linker 3 hu OKT3
9510 HM1.24 x CD3 hu C.HM1 .24 G236R/L328R Homo Linker 3 hu OKT3
951 1 CD19 x CD3 hu 4G7 G236R/L328R Homo Linker 3 hu OKT3
9550 HER2 x CD3 trastuzumab G236R/L328R Hetero TF-HA Linker 3 hu OKT3
9551 HM1.24 x CD3 hu C.HM1 .24 G236R/L328R Hetero TF-HA Linker 3 hu OKT3
9552 CD19 x CD3 hu 4G7 G236R/L328R Hetero TF-HA Linker 3 hu OKT3
8427 HER2 only trastuzumab Native lgG1 Homo None None
5627 HM1.24 only hu C.HM1 .24 Native lgG1 Homo None None
6166 HM1.24 only hu C.HM1 .24 G236R/L328R Homo None None a hu = humanized
b G236R/L328R is an Fc knockout variant that ablates binding to all FcyRs and complement, and ablates FcyR- and complement- mediated effector function.
c Homo = Homodimeric native IgG Fc region. Hetero TF-HA = Y349T/T394F and
S364H/F405A variant CH3 domains that favor heterodimerization and disfavor
homodimerization.
d Linker 3 = GGGGSGGGGSGGGGSGGGGSGGGGSGGGG (SEQ ID NO. 45)
Linker 4 = SSDKTHTSPPSPSG (SEQ ID NO. 46)
[275] The immunoglobulins in Table 5 were constructed as described above in the pTT5 expression vector. Heavy and light chains for each immunoglobulin were cotransfected into HEK293E cells for expression, and resulting protein was purified from the supernatant using protein A affinity chromatography, as described above. [276] The proteins were visualized electrophoretically using the Agilent Bioanalyzer as described above. Representative gels are shown in Figure 24. As expected, under reducing conditions three bands were observed at the approximate calculated molecular weights, corresponding to the light chain (-25 KDa) and two heavy chains (-65 KDa) for each mAb- Fv. Under non-reducing conditions a main band was observed for each immunoglobulin (-180 KDa) corresponding to the tetrameric mAb-Fv species (two heavy chains, each complexed with a light chain). The additional band in the non-reducing gels for 9550, 9551 , and 9552 is attributed to monomeric heavy chain / light chain pair (-90 KDa), similar to as was observed for the aCD19 x aFcyRllb mAb-Fv. Nonetheless, based on the band intensities of the aCD3 mAb-Fv's the heterodimeric heavy chain complexes are the dominant species for the 9550, 9551 , and 9552 constructs.
[277] The immunoglobulins were analyzed using SEC to measure their size and test the native-like behavior of the purified samples, as described above. Results for select mAb-Fv samples are shown in Figure 25. The mAb-Fv samples showed a main peak at an elution time that, based on a molecular weight standard (not shown), corresponded to the approximate molecular weight of the tetrameric mAb-Fv species (-180 KDa). The symmetry of the peaks and dominant population of the proper species for all of the aCD3 mAb-Fv's (aHer2, aCD19, and aHM1.24) indicated that the mAb-Fv format is robust for co- engagement of CD3 antigen. Consistent with the SDS-PAGE results, 9551 showed an additional smaller peak at a later elution time, attributed to the presence of monomeric heavy chain / light chain pairs.
[278] Affinity of the 9509 aHer2 x aCD3 mAb-Fv for CD3+ T cells was measured using a competition experiment with labeled OKT3 antibody. 9509 mAb-Fv at various concentrations was pre-incubated with -0.8 million PBMC on ice for 1 hour. Commercial FITC-labeled OKT3 was added to 50 ng/ml for 45 minutes. The binding buffer was 3% FBS/PBS, and samples were washed twice before fixing with 1 % Paraformaldehyde/PBS. Binding of OKT3- FITC to T cells was detected on a FACSCanto II flow cytometer (BD Biosciences) by analyzing CD16 D20" lymphocytes based on front scatter (FSS) and side scatter (SSC) scattering pattern that were negative for CD16 and CD20 using commercial labeled anti- CD16 and anti-CD20 antibody reagents. The results in Figure 26 demonstrate dose- dependent binding of the aHer2 x aCD3 mAb-Fv to T cells.
[279] Previous groups have demonstrated CD3-mediated cytotoxic killing by bispecific antibody fragments that co-engage CD3 and various target antigens (reviewed in Baeuerle & Reinhardt, 2009, Cancer Res 69[12]:4941-4). We tested the capacity of the CD3 co- engaging mAb-Fv's to mediate cytotoxic killing against tumor cells expressing Her2 and HM1 .24. Cytotoxicity assays were carried out as above, monitoring cell lysis by lactate dehydrogenase (LDH) release as described.
[280] aHer2 x aCD3 mAb-Fv's utilized purified T cells as effectors. T cells were purified using a T Cell Enrichment Kit from Stemcell Technologies (Vancouver, Canada). The 9509 aHer2 x aCD3 mAb-Fv was tested against Her2+ SK-BR-3 breast carcinoma cells at an effector to target ratio of 200,000 T cells to 10,000 target cells. Incubation time was 24 hrs. The 9551 aHM1 .24 x aCD3 mAb-Fv was used as a negative control. The results in Figure 27 show strong CD3-mediated cytotoxicity against the Her2+ target cells mediated by the Her2-directed mAb-Fv, while the HM1 .24-directed mAb-Fv showed no killing activity. The 9344 aHer2 x aCD3 mAb-Fv was tested against Her2+ SKOV3 ovarian carcinoma cells at an effector to target ratio of 250,000 T cells to 10,000 target cells. Incubation time was 24 hrs. Here the aHer2 lgG1 WT antibody trastuzumab was used as a negative control. The results in Figure 28 show strong CD3-mediated killing against the target cells by the mAb-Fv. In contrast, the WT lgG1 anti-Her2 antibody showed no capacity to mediate cytotoxic activity by the isolated T cells.
[281] The 9551 aHM1 .24 x aCD3 mAb-Fv was tested for CD3-mediated cytotoxic activity using purified PBMCs. Because this mAb-Fv contains the knockout variant Fc region G236R/L328R, it does not have the capacity to mediate FcyR-mediated cytotoxic activity by other immune cells in the heterogeneous PBMC mixture. Both knockout variant (6166) and WT lgG1 (5627) anti-HM1.24 antibodies were tested as controls. In this experiment HM1 .24+ RPMI 8224 myeloma cells were used as targets (seeded at 10,000 per well), and 500,000 PBMCs were added as effectors at a 50:1 effector/target cell ratio. Incubation time for this assay was 23 hrs. The results in Figure 29 show that the aHM1 .24 x aCD3 mAb-Fv mediated strong cytotoxic activity against the tumor cells. The 6166 Fc knockout anti- HMI .24 antibody was completely devoid of activity, consistent with it's lack of FcyR binding, and of course its inability as a native antibody to co-engage CD3. The 5627 WT lgG1 anti- HMI .24 antibody mediated very weak ADCC activity, if any at all, presumably due to FcyRllla-mediated killing by NK cells in the PBMC mixture. The dramatic CD3-mediated activity of the mAb-Fv relative to the FcyR-mediated activity of the normal lgG1 antibody targeting the same antigen demonstrates the potential of the novel formats herein for enabling powerful functional activities.
[282] All cited references are herein expressly incorporated by reference in their entirety.
[283] Whereas particular embodiments of the invention have been described above for purposes of illustration, it will be appreciated by those skilled in the art that numerous variations of the details may be made without departing from the invention as described in the appended claims.

Claims

CLAIMS We claim:
1. An antibody analog comprising:
a) a first heavy chain and an associated light chain, wherein said first heavy and light chains form a first antigen binding site, wherein the C-terminus of the CH3 domain of said first heavy chain is covalently attached to a C-terminal variable heavy domain;
b) a second heavy chain and an associated light chain, wherein said second heavy and light chains form a second antigen binding site, wherein the C-terminus of the CH3 domain of said second heavy chain is covalently attached to a C-terminal variable light domain;
such that said C-terminal variable heavy and light domains form a third antigen binding site.
2. An antibody analog according to claim 1 wherein two of said antigen binding sites are the same and the third is different.
3. An antibody analog according to claim 1 wherein the antigen bound by said third antigen binding site is CD3.
4. An antibody analog according to claim 1 wherein the antigen bound by said third antigen binding site is FcYRIIb.
5. An antibody analog according to claim 1 wherein all three of said antigen binding sites are the same.
6. An antibody analog according to claim 1 wherein said the C-terminus of said C-terminal variable heavy domain is covalently attached to a CH1 domain and wherein said C-terminus of said C-terminal variable light domain is covalently attached to a CL domain.
7. An antibody analog according to claim 1 or 6 wherein each heavy chain comprises at least one amino acid substitution within the Fc region.
8. An antibody analog according to claim 7 wherein each heavy chain comprises a different amino acid substitution, such that a heterodimeric Fc region is more stable than a homodimeric Fc region.
9. An antibody analog according to claim 7 wherein said substitution reduces binding to at least one FcyR receptor.
10. An antibody analog according to claim 7 wherein said amino acid substitution increases binding to at least one FcyR receptor.
1 1 . An antibody analog according to claim 7 wherein said amino acid substitution increases binding to the FcRn receptor.
12. An antibody analog according to claim 7 wherein said amino acid substitutions are selected from the group consisting of 349A, 349C, 349E, 349I, 349K, 349S, 349T, 349W, 351 E, 351 K, 354C, 356K, 357K, 364C, 364D, 364E, 364F, 364G, 364H, 364R, 364T, 364Y, 366D, 366K, 366S, 366W, 366Y, 368A, 368E, 368K, 368S, 370C, 370D, 370E, 370G, 370R, 370S, 370V, 392D, 392E, 394F, 394S, 394W, 394Y, 395T, 395V, 396T, 397E, 397S, 397T, 399K, 401 K, 405A, 405S, 407T, 407V, 409D, 409E, 41 1 D, 41 1 E, 41 1 K, and 439D.
13. An antibody analog comprising:
a) a first heavy chain comprising a variable heavy domain;
b) a second chain comprising the Fc domain of a heavy chain covalently linked to a light chain, wherein said first heavy chain and said second chain form a first antigen binding site; wherein the C-terminus of the CH3 domain of said first heavy chain is covalently attached to a variable heavy domain and the C-terminus of the CH3 domain of said second chain is covalently attached to a variable light domain, such that said variable heavy and light domains form a second antigen binding site.
14. An antibody analog according to claim 13 wherein a CH1 domain is covalently attached to said variable heavy domain covalently attached to the CH3 domain of said first heavy chain and wherein a CL domain is covalently attached to said variable light domain covalently attached to said CH3 domain of said second chain.
15. An antibody analog according to claim 13 or 14, wherein the wherein each of said protein chains comprises a variant CH3 domain relative to a native CH3 domain.
16. An antibody analog according to claim 15, wherein each of said variant CH3 domains comprise different amino acid substitutions, such that a heterodimeric protein pair is more stable than a homodimeric protein pair.
17. An antibody analog according to claim 16, wherein said substitution is selected from the group consisting of 349A, 349C, 349E, 349I, 349K, 349S, 349T, 349W, 351 E, 351 K, 354C, 356K, 357K, 364C, 364D, 364E, 364F, 364G, 364H, 364R, 364T, 364Y, 366D, 366K, 366S, 366W, 366Y, 368A, 368E, 368K, 368S, 370C, 370D, 370E, 370G, 370R, 370S, 370V, 392D, 392E, 394F, 394S, 394W, 394Y, 395T, 395V, 396T, 397E, 397S, 397T, 399K, 401 K, 405A, 405S, 407T, 407V, 409D, 409E, 41 1 D, 41 1 E, 41 1 K, and 439D, wherein numbering is according to the EU index as in Kabat.
PCT/US2010/047741 2009-09-02 2010-09-02 Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens WO2011028952A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23931609P 2009-09-02 2009-09-02
US61/239,316 2009-09-02

Publications (1)

Publication Number Publication Date
WO2011028952A1 true WO2011028952A1 (en) 2011-03-10

Family

ID=43086161

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/047741 WO2011028952A1 (en) 2009-09-02 2010-09-02 Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens

Country Status (2)

Country Link
US (3) US9493578B2 (en)
WO (1) WO2011028952A1 (en)

Cited By (188)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011091078A3 (en) * 2010-01-19 2011-12-01 Xencor, Inc. Antibody fc variants with enhanced complement activity
WO2011097527A3 (en) * 2010-02-04 2012-01-26 Xencor, Inc. Immunoprotection of therapeutic moieties using enhanced fc regions
WO2012058768A1 (en) * 2010-11-05 2012-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2013002362A1 (en) 2011-06-30 2013-01-03 中外製薬株式会社 Heterodimerized polypeptide
WO2013004842A2 (en) 2011-07-06 2013-01-10 Genmab A/S Antibody variants and uses thereof
WO2013033008A2 (en) 2011-08-26 2013-03-07 Merrimack Pharmaceuticals, Inc. Tandem fc bispecific antibodies
WO2013063702A1 (en) 2011-11-04 2013-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
WO2013187495A1 (en) 2012-06-14 2013-12-19 中外製薬株式会社 ANTIGEN-BINDING MOLECULE CONTAINING MODIFIED Fc REGION
WO2014006217A1 (en) 2012-07-06 2014-01-09 Genmab B.V. Dimeric protein with triple mutations
WO2014030728A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 Fcγriib-specific fc region variant
WO2014051433A1 (en) * 2012-09-27 2014-04-03 Merus B.V. BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS
WO2014104165A1 (en) 2012-12-27 2014-07-03 中外製薬株式会社 Heterodimerized polypeptide
WO2014108198A1 (en) 2013-01-10 2014-07-17 Genmab B.V. Human igg1 fc region variants and uses thereof
WO2014138449A1 (en) 2013-03-06 2014-09-12 Merrimack Pharmaceuticals, Inc. Anti-c-met tandem fc bispecific antibodies
WO2015001085A1 (en) 2013-07-05 2015-01-08 Genmab B.V. Humanized or chimeric cd3 antibodies
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
WO2015048272A1 (en) * 2013-09-25 2015-04-02 Amgen Inc. V-c-fc-v-c antibody
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
WO2015068847A1 (en) 2013-11-11 2015-05-14 中外製薬株式会社 Antigen-binding molecule containing modified antibody variable region
WO2016005593A1 (en) 2014-07-11 2016-01-14 Genmab A/S Antibodies binding axl
US9248182B2 (en) 2012-04-20 2016-02-02 Merus B.V. Methods and means for the production of Ig-like molecules
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
WO2016071376A3 (en) * 2014-11-06 2016-06-30 F. Hoffmann-La Roche Ag Fc-region variants with modified fcrn-binding and methods of use
US9382323B2 (en) 2009-04-02 2016-07-05 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
US9499605B2 (en) 2011-03-03 2016-11-22 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
WO2017009258A1 (en) 2015-07-10 2017-01-19 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
WO2017066878A1 (en) * 2015-10-22 2017-04-27 Canadian Blood Services Monovalent chimeras
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
WO2017086367A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Combination therapy using t cell redirection antigen binding molecule against cell having immunosuppressing function
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
WO2017159287A1 (en) 2016-03-14 2017-09-21 中外製薬株式会社 Cell injury inducing therapeutic drug for use in cancer therapy
US9771573B2 (en) 2012-10-03 2017-09-26 Zymeworks Inc. Methods of quantitating heavy and light chain polypeptide pairs
US9822186B2 (en) 2014-03-28 2017-11-21 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US9850320B2 (en) 2014-11-26 2017-12-26 Xencor, Inc. Heterodimeric antibodies to CD3 X CD20
US9856327B2 (en) 2014-11-26 2018-01-02 Xencor, Inc. Heterodimeric antibodies to CD3 X CD123
WO2018011421A1 (en) 2016-07-14 2018-01-18 Genmab A/S Multispecific antibodies against cd40 and cd137
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9914777B2 (en) 2015-07-10 2018-03-13 Merus N.V. Human CD3 binding antibody
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US9914776B2 (en) 2014-08-04 2018-03-13 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US9963513B2 (en) 2013-02-05 2018-05-08 Engmab Sàrl Method for the selection of antibodies against BCMA
WO2018083126A1 (en) 2016-11-01 2018-05-11 Genmab B.V. Polypeptide variants and uses thereof
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
WO2018146317A1 (en) 2017-02-10 2018-08-16 Genmab B.V. Polypeptide variants and uses thereof
WO2018162749A1 (en) 2017-03-09 2018-09-13 Genmab A/S Antibodies against pd-l1
US10077298B2 (en) 2012-11-28 2018-09-18 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US10087250B2 (en) 2012-10-08 2018-10-02 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
WO2018178396A1 (en) 2017-03-31 2018-10-04 Genmab Holding B.V. Bispecific anti-cd37 antibodies, monoclonal anti-cd37 antibodies and methods of use thereof
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
WO2018224609A1 (en) 2017-06-07 2018-12-13 Genmab B.V. Therapeutic antibodies based on mutated igg hexamers
US10155815B2 (en) 2013-02-26 2018-12-18 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10174124B2 (en) 2013-12-17 2019-01-08 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10179814B2 (en) 2014-07-17 2019-01-15 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
WO2019025545A1 (en) 2017-08-04 2019-02-07 Genmab A/S Binding agents binding to pd-l1 and cd137 and use thereof
WO2019032624A1 (en) * 2017-08-08 2019-02-14 Pionyr Immunotherapeutics, Inc. Compositions and methods for disabling meyloid cells expressing trem1
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US10287364B2 (en) 2013-03-15 2019-05-14 Xencor, Inc. Heterodimeric proteins
US10323094B2 (en) 2015-06-16 2019-06-18 Genentech, Inc. Humanized and affinity matured antibodies to FcRH5 and methods of use
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
WO2019131988A1 (en) 2017-12-28 2019-07-04 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
US10358479B2 (en) 2012-07-13 2019-07-23 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
WO2019145455A1 (en) 2018-01-24 2019-08-01 Genmab B.V. Polypeptide variants and uses thereof
WO2019155008A1 (en) 2018-02-09 2019-08-15 Genmab A/S Pharmaceutical compositions comprising bispecific antibodies directed against cd3 and cd20 and their uses
WO2019175198A2 (en) 2018-03-12 2019-09-19 Genmab A/S Antibodies
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
WO2019211472A1 (en) 2018-05-03 2019-11-07 Genmab B.V. Antibody variant combinations and uses thereof
US10472427B2 (en) 2013-01-14 2019-11-12 Xencor, Inc. Heterodimeric proteins
EP3486255A3 (en) * 2013-03-13 2019-11-13 Ibentrus Inc. Protein in which electrical interaction is introduced within hydrophobic interaction site and preparation method therefor
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
WO2019232484A1 (en) 2018-06-01 2019-12-05 Compugen Ltd Anti-pvrig/anti-tigit bispecific antibodies and methods of use
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
US10501545B2 (en) 2015-06-16 2019-12-10 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
WO2019243636A1 (en) 2018-06-22 2019-12-26 Genmab Holding B.V. Anti-cd37 antibodies and anti-cd20 antibodies, compositions and methods of use thereof
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10526417B2 (en) 2014-11-26 2020-01-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
WO2020012038A1 (en) 2018-07-13 2020-01-16 Genmab A/S Trogocytosis-mediated therapy using cd38 antibodies
WO2020012036A1 (en) 2018-07-13 2020-01-16 Genmab A/S Variants of cd38 antibody and uses thereof
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10544220B2 (en) 2015-01-08 2020-01-28 Genmab A/S Bispecific antibodies against CD3 and CD20
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
EP3632462A1 (en) 2012-07-06 2020-04-08 Genmab B.V. Dimeric protein with triple mutations
WO2020070313A1 (en) 2018-10-04 2020-04-09 Genmab Holding B.V. Pharmaceutical compositions comprising bispecific anti-cd37 antibodies
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
WO2020094744A1 (en) 2018-11-06 2020-05-14 Genmab A/S Antibody formulation
US10738133B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10774156B2 (en) 2015-01-22 2020-09-15 Eli Lilly And Company IgG bispecific antibodies and processes for preparation
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
WO2020225456A1 (en) 2019-05-09 2020-11-12 Genmab B.V. Dosage regimens for a combination of anti-dr5 antibodies for use in treating cancer
US10836828B2 (en) 2019-02-06 2020-11-17 Pionyr Immunotherapeutics, Inc. Anti-TREM1 antibodies and related methods
US10844127B2 (en) 2014-02-28 2020-11-24 Merus N.V. Antibodies that bind EGFR and ErbB3
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
WO2020246563A1 (en) 2019-06-05 2020-12-10 中外製薬株式会社 Antibody cleavage site-binding molecule
US10882918B2 (en) 2016-09-30 2021-01-05 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
WO2021006328A1 (en) 2019-07-10 2021-01-14 Chugai Seiyaku Kabushiki Kaisha Claudin-6 binding molecules and uses thereof
WO2021028587A1 (en) 2019-08-15 2021-02-18 Genmab A/S Pharmaceutical compositions comprising bispecific antibodies directed against cd3 and cd20 and their uses
US10934571B2 (en) 2002-07-18 2021-03-02 Merus N.V. Recombinant production of mixtures of antibodies
US10947319B2 (en) 2013-11-27 2021-03-16 Zymeworks Inc. Bispecific antigen-binding constructs targeting HER2
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
WO2021089850A1 (en) 2019-11-06 2021-05-14 Genmab B.V. Antibody variant combinations and uses thereof
US11007267B2 (en) 2015-06-16 2021-05-18 Genentech, Inc. Anti-CD3 antibodies and methods of use
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
WO2021131021A1 (en) 2019-12-27 2021-07-01 中外製薬株式会社 Anti-ctla-4 antibody and use thereof
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
WO2021144457A1 (en) 2020-01-16 2021-07-22 Genmab A/S Formulations of cd38 antibodies and uses thereof
US11084877B2 (en) 2014-09-12 2021-08-10 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
WO2021156258A1 (en) 2020-02-04 2021-08-12 BioNTech SE Treatment involving antigen vaccination and binding agents binding to pd-l1 and cd137
WO2021185934A1 (en) 2020-03-18 2021-09-23 Genmab A/S Antibodies binding to b7h4
WO2021201087A1 (en) 2020-03-31 2021-10-07 Chugai Seiyaku Kabushiki Kaisha Method for producing multispecific antigen-binding molecules
WO2021200857A1 (en) 2020-03-30 2021-10-07 国立大学法人三重大学 Bispecific antibody
US11155618B2 (en) 2018-04-02 2021-10-26 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
WO2021224499A1 (en) 2020-05-08 2021-11-11 Genmab A/S Bispecific antibodies against cd3 and cd20
US11225521B2 (en) 2016-06-28 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
WO2022018294A1 (en) 2020-07-23 2022-01-27 Genmab B.V. A combination of anti-dr5 antibodies and an immunomodulatory imide drug for use in treating multiple myeloma
US11236170B2 (en) 2016-06-14 2022-02-01 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
WO2022025220A1 (en) 2020-07-31 2022-02-03 中外製薬株式会社 Pharmaceutical composition including cell expressing chimeric receptor
US11242390B2 (en) 2016-03-22 2022-02-08 Hoffmann-La Roche Inc. Protease-activated T cell bispecific molecules
WO2022029011A1 (en) 2020-08-06 2022-02-10 BioNTech SE Binding agents for coronavirus s protein
WO2022045276A1 (en) 2020-08-28 2022-03-03 中外製薬株式会社 Heterodimer fc polypeptide
WO2022049220A2 (en) 2020-09-02 2022-03-10 Genmab A/S Antibody therapy
US11279770B2 (en) 2014-02-28 2022-03-22 Merus N.V. Antibody that binds ErbB-2 and ErbB-3
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
WO2022069724A1 (en) 2020-10-02 2022-04-07 Genmab A/S Antibodies capable of binding to ror2 and bispecific antibodies binding to ror2 and cd3
US11306156B2 (en) 2014-05-28 2022-04-19 Zymeworks Inc. Modified antigen binding polypeptide constructs and uses thereof
US11312770B2 (en) 2017-11-08 2022-04-26 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-PD-1 sequences
US11319355B2 (en) 2017-12-19 2022-05-03 Xencor, Inc. Engineered IL-2 Fc fusion proteins
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
US11359015B2 (en) 2015-07-15 2022-06-14 Genmab A/S Humanized or chimeric CD3 antibodies
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
WO2022189667A1 (en) 2021-03-12 2022-09-15 Genmab A/S Non-activating antibody variants
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US11466094B2 (en) 2016-11-15 2022-10-11 Genentech, Inc. Dosing for treatment with anti-CD20/anti-CD3 bispecific antibodies
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
KR20220142441A (en) 2020-02-14 2022-10-21 쿄와 기린 가부시키가이샤 Bispecific antibody that binds to CD3
WO2022234146A1 (en) 2021-05-07 2022-11-10 Genmab A/S PHARMACEUTICAL COMPOSITIONS COMPRISING BISPECIFIC ANTIBODIES BINDING TO B7H4 and CD3
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US11535679B2 (en) 2020-09-10 2022-12-27 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating follicular lymphoma
WO2022270612A1 (en) 2021-06-25 2022-12-29 中外製薬株式会社 Use of anti-ctla-4 antibody
WO2022270611A1 (en) 2021-06-25 2022-12-29 中外製薬株式会社 Anti–ctla-4 antibody
WO2022268740A1 (en) 2021-06-21 2022-12-29 Genmab A/S Combination dosage regime of cd137 and pd-l1 binding agents
US11548952B2 (en) 2020-09-10 2023-01-10 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating diffuse large B-cell lymphoma
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
WO2023031473A1 (en) 2021-09-06 2023-03-09 Genmab B.V. Antibodies capable of binding to cd27, variants thereof and uses thereof
US11608383B2 (en) 2020-09-10 2023-03-21 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating follicular lymphoma
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
WO2023057571A1 (en) 2021-10-08 2023-04-13 Genmab A/S Antibodies binding to cd30 and cd3
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
WO2023174952A1 (en) 2022-03-15 2023-09-21 Genmab A/S Binding agents binding to epcam and/or cd137
US11773170B2 (en) 2017-08-09 2023-10-03 Merus N.V. Antibodies that bind EGFR and cMET
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19
US11780925B2 (en) 2017-03-31 2023-10-10 Merus N.V. ErbB-2 and ErbB3 binding bispecific antibodies for use in the treatment of cells that have an NRG1 fusion gene
WO2023198839A2 (en) 2022-04-13 2023-10-19 Genmab A/S Bispecific antibodies against cd3 and cd20
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023218051A1 (en) 2022-05-12 2023-11-16 Genmab A/S Binding agents capable of binding to cd27 in combination therapy
WO2023218046A1 (en) 2022-05-12 2023-11-16 Genmab A/S Binding agents capable of binding to cd27 in combination therapy
US11845805B2 (en) 2020-09-10 2023-12-19 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating diffuse large B-cell lymphoma
US11845799B2 (en) 2019-12-13 2023-12-19 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
RU2810540C2 (en) * 2011-11-04 2023-12-27 Займворкс Бк Инк. DEVELOPMENT OF STABLE HETERODIMERIC ANTIBODY WITH MUTATIONS IN Fc DOMAIN
US11858995B2 (en) 2020-09-10 2024-01-02 Genmab A/S Bispecific antibodies against CD3 and CD20 for treating chronic lymphocytic leukemia
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
US11939394B2 (en) 2015-10-23 2024-03-26 Merus N.V. Binding molecules that inhibit cancer growth

Families Citing this family (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
CA2781539C (en) * 2009-11-23 2021-07-20 Amgen Inc. Monomeric antibody fc
US10053513B2 (en) * 2009-11-30 2018-08-21 Janssen Biotech, Inc. Antibody Fc mutants with ablated effector functions
KR101791430B1 (en) 2009-11-30 2017-10-30 얀센 바이오테크 인코포레이티드 ANTIBODY Fc MUTANTS WITH ABLATED EFFECTOR FUNCTIONS
EP3029066B1 (en) 2010-07-29 2019-02-20 Xencor, Inc. Antibodies with modified isoelectric points
WO2012069466A1 (en) * 2010-11-24 2012-05-31 Novartis Ag Multispecific molecules
TWI588156B (en) * 2011-03-28 2017-06-21 賽諾菲公司 Dual variable region antibody-like binding proteins having cross-over binding region orientation
EP2710042A2 (en) 2011-05-16 2014-03-26 Fabion Pharmaceuticals, Inc. Multi-specific fab fusion proteins and methods of use
WO2013055809A1 (en) * 2011-10-10 2013-04-18 Xencor, Inc. A method for purifying antibodies
TR201809151T4 (en) * 2011-10-11 2018-07-23 Hoffmann La Roche Improved binding of bispecific antibodies.
WO2013188693A1 (en) 2012-06-15 2013-12-19 Imaginab, Inc. Antigen binding constructs to cd3
WO2014084607A1 (en) * 2012-11-27 2014-06-05 아주대학교산학협력단 Ch3 domain variant pair inducing formation of heterodimer of heavy chain constant region of antibody at high efficiency, method for preparing same, and use thereof
US20140154255A1 (en) * 2012-11-30 2014-06-05 Abbvie Biotherapeutics Inc. Anti-vegf antibodies and their uses
EP2945969A1 (en) 2013-01-15 2015-11-25 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
DK3587448T3 (en) 2013-03-15 2021-08-09 Xencor Inc HETERODIMER PROTEINS
JP6618893B2 (en) 2013-04-29 2019-12-11 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Asymmetric antibodies with altered FC receptor binding and methods of use
PE20151807A1 (en) 2013-04-29 2015-12-02 Hoffmann La Roche MODIFIED ANTIBODIES OF BINDING TO HUMAN FCRN AND METHOD OF USE
MX2016003292A (en) 2013-09-13 2016-06-24 Beigene Ltd Anti-pd1 antibodies and their use as therapeutics and diagnostics.
EP3757130A1 (en) 2013-09-26 2020-12-30 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
CA2930307A1 (en) * 2013-11-13 2015-05-21 Zymeworks Inc. Monovalent antigen binding constructs targeting egfr and/or her2 and uses thereof
CA2932364A1 (en) 2014-01-15 2015-07-23 F. Hoffmann-La Roche Ag Fc-region variants with improved protein a-binding
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
JOP20200096A1 (en) 2014-01-31 2017-06-16 Children’S Medical Center Corp Antibody molecules to tim-3 and uses thereof
WO2015153765A1 (en) 2014-04-01 2015-10-08 Adimab, Llc Multispecific antibody analogs comprising a common light chain, and methods of their preparation and use
WO2016001810A1 (en) 2014-07-01 2016-01-07 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
JP6526189B2 (en) 2014-07-03 2019-06-05 ベイジーン リミテッド Anti-PD-L1 antibodies and their use for therapy and diagnosis
ES2905301T3 (en) 2014-07-22 2022-04-07 Apollomics Inc Anti-PD-1 antibodies
KR102476226B1 (en) 2014-08-05 2022-12-12 아폴로믹스 인코포레이티드 Anti-pd-l1 antibodies
EP3191127A1 (en) 2014-09-13 2017-07-19 Novartis AG Combination therapies of egfr inhibitors
US20160176969A1 (en) 2014-11-26 2016-06-23 Xencor, Inc. Heterodimeric antibodies including binding to cd8
CN108472360B (en) 2015-04-10 2023-01-17 阿迪马布有限责任公司 Method for purifying heterodimeric multispecific antibodies from parent homodimeric antibody species
CN115960230A (en) 2015-08-07 2023-04-14 伊麦吉纳博公司 Antigen binding constructs for targeting molecules
EP3307321A4 (en) 2015-08-26 2019-04-17 Bison Therapeutics Inc. Multispecific antibody platform and related methods
MA43348A (en) 2015-10-01 2018-08-08 Novo Nordisk As PROTEIN CONJUGATES
KR101851380B1 (en) 2015-10-12 2018-04-23 아주대학교산학협력단 Method to generate CH3 domain mutant pairs of heterodimeric Fc using yeast mating and CH3 domain mutant pairs thereby
WO2017157305A1 (en) 2016-03-15 2017-09-21 Generon (Shanghai) Corporation Ltd. Multispecific fab fusion proteins and use thereof
US11291721B2 (en) 2016-03-21 2022-04-05 Marengo Therapeutics, Inc. Multispecific and multifunctional molecules and uses thereof
NZ749997A (en) 2016-07-05 2022-11-25 Beigene Ltd Combination of a pd-l antagonist and a raf inhibitor for treating cancer
EP3511340A4 (en) 2016-08-10 2020-03-18 Ajou University Industry-Academic Cooperation Foundation Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same
DK3500299T3 (en) 2016-08-19 2024-01-29 Beigene Switzerland Gmbh Combination of zanubrutinib with an anti-CD20 or an anti-PD-1 antibody for use in the treatment of cancer
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
EP3573989A4 (en) 2017-01-25 2020-11-18 Beigene, Ltd. Crystalline forms of (s) -7- (1- (but-2-ynoyl) piperidin-4-yl) -2- (4-phenoxyphenyl) -4, 5, 6, 7-tetrahy dropyrazolo [1, 5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
WO2018144784A1 (en) 2017-02-01 2018-08-09 Smet Pharmaceutical Inc. MONOMERIC HUMAN IgG1 Fc AND BISPECIFIC ANTIBODIES
JOP20190189A1 (en) 2017-02-02 2019-08-01 Amgen Res Munich Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies
MX2019009468A (en) 2017-02-08 2020-01-20 Dragonfly Therapeutics Inc Multi-specific binding proteins for activation of natural killer cells and therapeutic uses thereof to treat cancer.
US20200291089A1 (en) 2017-02-16 2020-09-17 Elstar Therapeutics, Inc. Multifunctional molecules comprising a trimeric ligand and uses thereof
EP4273258A3 (en) 2017-02-20 2024-01-17 Dragonfly Therapeutics, Inc. Proteins binding her2, nkg2d and cd16
WO2018156735A1 (en) * 2017-02-22 2018-08-30 H. Lee Moffitt Cancer Center And Research Institute, Inc. Bispecific antibody for cancer immunotherapy
EP3606560A2 (en) * 2017-04-05 2020-02-12 Novo Nordisk A/S Oligomer extended insulin-fc conjugates
WO2018222901A1 (en) 2017-05-31 2018-12-06 Elstar Therapeutics, Inc. Multispecific molecules that bind to myeloproliferative leukemia (mpl) protein and uses thereof
EP3645569A4 (en) 2017-06-26 2021-03-24 BeiGene, Ltd. Immunotherapy for hepatocellular carcinoma
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
CN111801334B (en) 2017-11-29 2023-06-09 百济神州瑞士有限责任公司 Treatment of indolent or invasive B-cell lymphomas using combinations comprising BTK inhibitors
JP2021512630A (en) 2018-02-08 2021-05-20 ドラゴンフライ セラピューティクス, インコーポレイテッド Antibody variable domain targeting NKG2D receptor
WO2019178362A1 (en) 2018-03-14 2019-09-19 Elstar Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
US20210009711A1 (en) 2018-03-14 2021-01-14 Elstar Therapeutics, Inc. Multifunctional molecules and uses thereof
US10633458B2 (en) 2018-04-10 2020-04-28 Y-Biologics Inc. Cell engaging binding molecules
PE20210320A1 (en) 2018-06-01 2021-02-16 Novartis Ag BINDING MOLECULES AGAINST BCMA AND THE USES OF THEM
CN112584859A (en) 2018-07-02 2021-03-30 美国安进公司 anti-STEAP 1 antigen binding proteins
JP2021530246A (en) 2018-07-03 2021-11-11 マレンゴ・セラピューティクス,インコーポレーテッド Anti-TCR antibody molecule and its use
US11377477B2 (en) 2018-10-12 2022-07-05 Xencor, Inc. PD-1 targeted IL-15/IL-15RALPHA fc fusion proteins and uses in combination therapies thereof
WO2020086758A1 (en) 2018-10-23 2020-04-30 Dragonfly Therapeutics, Inc. Heterodimeric fc-fused proteins
US11618776B2 (en) 2018-12-20 2023-04-04 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
CN114127113A (en) 2019-02-21 2022-03-01 马伦戈治疗公司 Multifunctional molecules binding to calreticulin and uses thereof
SG11202109061YA (en) 2019-02-21 2021-09-29 Marengo Therapeutics Inc Multifunctional molecules that bind to t cell related cancer cells and uses thereof
SG11202109122SA (en) 2019-02-21 2021-09-29 Marengo Therapeutics Inc Anti-tcr antibody molecules and uses thereof
CA3130628A1 (en) 2019-02-21 2020-08-27 Marengo Therapeutics, Inc. Multifunctional molecules that bind to t cells and uses thereof to treat autoimmune disorders
AU2020224681A1 (en) 2019-02-21 2021-09-16 Marengo Therapeutics, Inc. Antibody molecules that bind to NKp30 and uses thereof
TW202128757A (en) 2019-10-11 2021-08-01 美商建南德克公司 Pd-1 targeted il-15/il-15ralpha fc fusion proteins with improved properties
WO2021138407A2 (en) 2020-01-03 2021-07-08 Marengo Therapeutics, Inc. Multifunctional molecules that bind to cd33 and uses thereof
KR20220145854A (en) * 2020-02-27 2022-10-31 페인스 테라퓨틱스 인코포레이티드 Antibodies conjugated to fatty acid molecules and uses thereof
CA3180321A1 (en) 2020-04-24 2021-10-28 Marengo Therapeutics, Inc. Multifunctional molecules that bind to t cell related cancer cells and uses thereof
JP2023539645A (en) 2020-08-26 2023-09-15 マレンゴ・セラピューティクス,インコーポレーテッド Antibody molecules that bind to NKP30 and uses thereof
AU2021333779A1 (en) 2020-08-26 2023-04-13 Marengo Therapeutics, Inc. Methods of detecting TRBC1 or TRBC2
EP4204450A2 (en) 2020-08-26 2023-07-05 Marengo Therapeutics, Inc. Multifunctional molecules that bind to calreticulin and uses thereof
WO2022216993A2 (en) 2021-04-08 2022-10-13 Marengo Therapeutics, Inc. Multifuntional molecules binding to tcr and uses thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992011018A1 (en) 1990-12-19 1992-07-09 Protein Design Labs, Inc. Improved humanized immunoglobulins
WO1995009917A1 (en) * 1993-10-07 1995-04-13 The Regents Of The University Of California Genetically engineered bispecific tetravalent antibodies
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US20060134150A1 (en) 2000-12-22 2006-06-22 Jane Werling Submicron suspensions with polymorph control
WO2010028796A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Trispecific hexavalent antibodies
WO2010034441A1 (en) * 2008-09-26 2010-04-01 F. Hoffmann-La Roche Ag Bispecific anti-egfr/anti-igf-1r antibodies
WO2010112193A1 (en) * 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
WO2010115552A1 (en) * 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-3/anti-c-met antibodies
WO2010115589A1 (en) * 2009-04-07 2010-10-14 Roche Glycart Ag Trivalent, bispecific antibodies
WO2010115551A1 (en) * 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies

Family Cites Families (490)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
CU22545A1 (en) 1994-11-18 1999-03-31 Centro Inmunologia Molecular OBTAINING A CHEMICAL AND HUMANIZED ANTIBODY AGAINST THE RECEPTOR OF THE EPIDERMAL GROWTH FACTOR FOR DIAGNOSTIC AND THERAPEUTIC USE
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4169888A (en) 1977-10-17 1979-10-02 The Upjohn Company Composition of matter and process
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4256746A (en) 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
JPS55102583A (en) 1979-01-31 1980-08-05 Takeda Chem Ind Ltd 20-acyloxy-20-demethylmaytansinoid compound
JPS55162791A (en) 1979-06-05 1980-12-18 Takeda Chem Ind Ltd Antibiotic c-15003pnd and its preparation
JPS6023084B2 (en) 1979-07-11 1985-06-05 味の素株式会社 blood substitute
JPS5645483A (en) 1979-09-19 1981-04-25 Takeda Chem Ind Ltd C-15003phm and its preparation
JPS5645485A (en) 1979-09-21 1981-04-25 Takeda Chem Ind Ltd Production of c-15003pnd
EP0028683A1 (en) 1979-09-21 1981-05-20 Takeda Chemical Industries, Ltd. Antibiotic C-15003 PHO and production thereof
WO1981001145A1 (en) 1979-10-18 1981-04-30 Univ Illinois Hydrolytic enzyme-activatible pro-drugs
US4364935A (en) 1979-12-04 1982-12-21 Ortho Pharmaceutical Corporation Monoclonal antibody to a human prothymocyte antigen and methods of preparing same
WO1982001188A1 (en) 1980-10-08 1982-04-15 Takeda Chemical Industries Ltd 4,5-deoxymaytansinoide compounds and process for preparing same
US4450254A (en) 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4313946A (en) 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4315929A (en) 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
JPS57192389A (en) 1981-05-20 1982-11-26 Takeda Chem Ind Ltd Novel maytansinoid
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4753894A (en) 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US4943533A (en) 1984-03-01 1990-07-24 The Regents Of The University Of California Hybrid cell lines that produce monoclonal antibodies to epidermal growth factor receptor
US4970198A (en) 1985-10-17 1990-11-13 American Cyanamid Company Antitumor antibiotics (LL-E33288 complex)
DE3675588D1 (en) 1985-06-19 1990-12-20 Ajinomoto Kk HAEMOGLOBIN TIED TO A POLY (ALKENYLENE OXIDE).
WO1987005330A1 (en) 1986-03-07 1987-09-11 Michel Louis Eugene Bergh Method for enhancing glycoprotein stability
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
EP0271581B1 (en) 1986-04-17 1993-01-13 Kyowa Hakko Kogyo Co., Ltd. Novel compounds dc-88a and dc-89a1 and process for their preparation
NZ220504A (en) 1986-05-29 1991-09-25 Univ Melbourne Fc receptor site an immunoglobulin and dna sequences encodng it
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US4880935A (en) 1986-07-11 1989-11-14 Icrf (Patents) Limited Heterobifunctional linking agents derived from N-succinimido-dithio-alpha methyl-methylene-benzoates
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
GB8705477D0 (en) 1987-03-09 1987-04-15 Carlton Med Prod Drug delivery systems
WO1988007089A1 (en) 1987-03-18 1988-09-22 Medical Research Council Altered antibodies
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
PT88641B (en) 1987-10-02 1993-04-30 Genentech Inc METHOD FOR PREPARING A VARIETY OF ADHESION
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
US5053394A (en) 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
IL106992A (en) 1988-02-11 1994-06-24 Bristol Myers Squibb Co Acylhydrazone derivatives of anthracycline and methods for their preparation
US5084468A (en) 1988-08-11 1992-01-28 Kyowa Hakko Kogyo Co., Ltd. Dc-88a derivatives
US5576184A (en) 1988-09-06 1996-11-19 Xoma Corporation Production of chimeric mouse-human antibodies with specificity to human tumor antigens
US5681566A (en) 1988-10-24 1997-10-28 3I Research Exploitation Limited Antibody conjugates with two or more covalently linked FC regions
US20040049014A1 (en) 1988-12-28 2004-03-11 Protein Design Labs, Inc. Humanized immunoglobulins
JP2598116B2 (en) 1988-12-28 1997-04-09 協和醗酵工業株式会社 New substance DC113
US5266491A (en) 1989-03-14 1993-11-30 Mochida Pharmaceutical Co., Ltd. DNA fragment and expression plasmid containing the DNA fragment
JP3051411B2 (en) 1989-03-14 2000-06-12 持田製薬株式会社 Novel DNA and expression plasmid containing it
US5187186A (en) 1989-07-03 1993-02-16 Kyowa Hakko Kogyo Co., Ltd. Pyrroloindole derivatives
JP2510335B2 (en) 1989-07-03 1996-06-26 協和醗酵工業株式会社 DC-88A derivative
GB8923123D0 (en) 1989-10-13 1989-11-29 Connaught Lab A vaccine for human immunodeficiency virus
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
CA2026147C (en) 1989-10-25 2006-02-07 Ravi J. Chari Cytotoxic agents comprising maytansinoids and their therapeutic use
JPH04505709A (en) 1989-11-07 1992-10-08 ブリストル―マイアーズ スクイブ カンパニー oligomeric immunoglobulin
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
EP0530225A4 (en) 1990-05-07 1993-09-22 Scripps Clinic And Research Foundation Intermediates in the formation of the calicheamicin and esperamicin oligosaccharides
WO1991019515A1 (en) 1990-06-21 1991-12-26 The Board Of Trustees Of The Leland Stanford Junior University Oligomeric immunoglobulin constant domain mutant with enhanced complement-mediated cytolytic activity
US5198342A (en) 1990-07-05 1993-03-30 Immunex Corporation DNA encoding IgA Fc receptors
EP0547137A4 (en) 1990-08-31 1993-12-08 Bristol-Myers Squibb Company Homoconjugated immunoglobulins
US5968509A (en) 1990-10-05 1999-10-19 Btp International Limited Antibodies with binding affinity for the CD3 antigen
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
CA2082160C (en) 1991-03-06 2003-05-06 Mary M. Bendig Humanised and chimeric monoclonal antibodies
GB9105245D0 (en) 1991-03-12 1991-04-24 Lynxvale Ltd Binding molecules
US6797492B2 (en) 1991-05-17 2004-09-28 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
WO1992022324A1 (en) 1991-06-14 1992-12-23 Xoma Corporation Microbially-produced antibody fragments and their conjugates
WO1992022653A1 (en) 1991-06-14 1992-12-23 Genentech, Inc. Method for making humanized antibodies
US5264586A (en) 1991-07-17 1993-11-23 The Scripps Research Institute Analogs of calicheamicin gamma1I, method of making and using the same
IE922437A1 (en) 1991-07-25 1993-01-27 Idec Pharma Corp Recombinant antibodies for human therapy
US6329509B1 (en) 1991-08-14 2001-12-11 Genentech, Inc. Anti-IgE antibodies
EP0604580A1 (en) 1991-09-19 1994-07-06 Genentech, Inc. EXPRESSION IN E. COLI OF ANTIBODY FRAGMENTS HAVING AT LEAST A CYSTEINE PRESENT AS A FREE THIOL, USE FOR THE PRODUCTION OF BIFUNCTIONAL F(ab') 2? ANTIBODIES
US5623053A (en) 1992-01-10 1997-04-22 California Institute Of Technology Soluble mammal-derived Fc receptor which binds at a pH ranging from about 5.5 to 6.5 and releases at a pH ranging from about 7.5 to 8.5
US5622929A (en) 1992-01-23 1997-04-22 Bristol-Myers Squibb Company Thioether conjugates
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
GB9206422D0 (en) 1992-03-24 1992-05-06 Bolt Sarah L Antibody preparation
CA2076465C (en) 1992-03-25 2002-11-26 Ravi V. J. Chari Cell binding agent conjugates of analogues and derivatives of cc-1065
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
CA2118508A1 (en) 1992-04-24 1993-11-11 Elizabeth S. Ward Recombinant production of immunoglobulin-like domains in prokaryotic cells
US6329507B1 (en) 1992-08-21 2001-12-11 The Dow Chemical Company Dimer and multimer forms of single chain polypeptides
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
PL174721B1 (en) 1992-11-13 1998-09-30 Idec Pharma Corp Monoclonal antibody anty-cd2
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
DK0672142T3 (en) 1992-12-04 2001-06-18 Medical Res Council Multivalent and multi-specific binding proteins as well as their preparation and use
CA2117477C (en) 1992-12-11 2001-06-12 Peter S. Mezes Multivalent single chain antibodies
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US20030108548A1 (en) 1993-06-01 2003-06-12 Bluestone Jeffrey A. Methods and materials for modulation of the immunosuppressive activity and toxicity of monoclonal antibodies
US5885573A (en) 1993-06-01 1999-03-23 Arch Development Corporation Methods and materials for modulation of the immunosuppressive activity and toxicity of monoclonal antibodies
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
GB9316989D0 (en) 1993-08-16 1993-09-29 Lynxvale Ltd Binding molecules
CA2173150C (en) 1993-10-01 2004-11-30 Kyoichi Sakakibara Novel peptide derivatives
GB9401182D0 (en) 1994-01-21 1994-03-16 Inst Of Cancer The Research Antibodies to EGF receptor and their antitumour effect
JPH09512705A (en) 1994-03-29 1997-12-22 セルテック セラピューティクス リミテッド Antibodies to E-selectin
US5641780A (en) 1994-04-22 1997-06-24 Kyowa Hakko Kogyo Co., Ltd. Pyrrolo-indole derivatives
JPH07309761A (en) 1994-05-20 1995-11-28 Kyowa Hakko Kogyo Co Ltd Method for stabilizing duocamycin derivative
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5945311A (en) 1994-06-03 1999-08-31 GSF--Forschungszentrumfur Umweltund Gesundheit Method for producing heterologous bi-specific antibodies
US5798100A (en) * 1994-07-06 1998-08-25 Immunomedics, Inc. Multi-stage cascade boosting vaccine
US5550246A (en) 1994-09-07 1996-08-27 The Scripps Research Institute Calicheamicin mimics
US5541087A (en) 1994-09-14 1996-07-30 Fuji Immunopharmaceuticals Corporation Expression and export technology of proteins as immunofusins
US5804396A (en) 1994-10-12 1998-09-08 Sugen, Inc. Assay for agents active in proliferative disorders
US6214388B1 (en) 1994-11-09 2001-04-10 The Regents Of The University Of California Immunoliposomes that optimize internalization into target cells
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US6030613A (en) 1995-01-17 2000-02-29 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
US6485726B1 (en) 1995-01-17 2002-11-26 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
EP0805628B1 (en) 1995-01-17 2003-05-02 Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of immunogens
US6086875A (en) 1995-01-17 2000-07-11 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of immunogens
US6037453A (en) 1995-03-15 2000-03-14 Genentech, Inc. Immunoglobulin variants
EP3103799B1 (en) 1995-03-30 2018-06-06 OSI Pharmaceuticals, LLC Quinazoline derivatives
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
GB9508565D0 (en) 1995-04-27 1995-06-14 Zeneca Ltd Quiazoline derivative
US6444789B1 (en) 1995-05-03 2002-09-03 Applied Research Systems Ars Holding N.V. CD16-II variants
CA2222231A1 (en) 1995-06-07 1996-12-19 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth of tumors
US6365161B1 (en) 1995-06-07 2002-04-02 Medarex, Inc. Therapeutic compounds comprised of anti-FC receptor binding agents
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US7696338B2 (en) 1995-10-30 2010-04-13 The United States Of America As Represented By The Department Of Health And Human Services Immunotoxin fusion proteins and means for expression thereof
US5790396A (en) 1995-12-19 1998-08-04 Kabushiki Kaisha Toshiba Neutral point clamped (NPC) inverter control system
EP0871490B1 (en) 1995-12-22 2003-03-19 Bristol-Myers Squibb Company Branched hydrazone linkers
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
EP0904107B1 (en) 1996-03-18 2004-10-20 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
US6254868B1 (en) 1996-03-20 2001-07-03 Immunomedics, Inc. Glycosylated humanized B-cell specific antibodies
EA001595B1 (en) 1996-04-12 2001-06-25 Варнер-Ламберт Компани Irreversible inhibitors of tyrosine kinases.
WO1997043316A1 (en) 1996-05-10 1997-11-20 Beth Israel Deaconess Medical Center, Inc. Physiologically active molecules with extended half-lives and methods of using same
US5834597A (en) 1996-05-20 1998-11-10 Protein Design Labs, Inc. Mutated nonactivating IgG2 domains and anti CD3 antibodies incorporating the same
ATE253593T1 (en) 1996-07-16 2003-11-15 Plueckthun Andreas Prof Dr IMMUNOGLOBULIN SUPERFAMILY DOMAINS AND FRAGMENTS WITH INCREASED SOLUBILITY
WO1998005787A1 (en) 1996-08-02 1998-02-12 Bristol-Myers Squibb Company A method for inhibiting immunoglobulin-induced toxicity resulting from the use of immunoglobulins in therapy and in vivo diagnosis
WO1998023289A1 (en) 1996-11-27 1998-06-04 The General Hospital Corporation MODULATION OF IgG BINDING TO FcRn
WO1998031806A2 (en) 1997-01-21 1998-07-23 Human Genome Sciences, Inc. Fc RECEPTORS AND POLYPEPTIDES
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
UA73073C2 (en) 1997-04-03 2005-06-15 Уайт Холдінгз Корпорейшн Substituted 3-cyan chinolines
EP1255209A3 (en) 1997-04-11 2009-02-11 California Institute Of Technology Apparatus and method for automated protein design
WO1998047089A1 (en) 1997-04-11 1998-10-22 California Institute Of Technology Apparatus and method for automated protein design
US7951917B1 (en) * 1997-05-02 2011-05-31 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US20020062010A1 (en) 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
JP2002512624A (en) 1997-05-21 2002-04-23 バイオベーション リミテッド Method for producing non-immunogenic protein
GB9712892D0 (en) 1997-06-20 1997-08-20 Eclagen Ltd Identification of mhc binding peptides
US20040191256A1 (en) 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
ZA986732B (en) 1997-07-29 1999-02-02 Warner Lambert Co Irreversible inhibitiors of tyrosine kinases
ZA986729B (en) 1997-07-29 1999-02-02 Warner Lambert Co Irreversible inhibitors of tyrosine kinases
TW436485B (en) 1997-08-01 2001-05-28 American Cyanamid Co Substituted quinazoline derivatives
US20030105294A1 (en) 1998-02-25 2003-06-05 Stephen Gillies Enhancing the circulating half life of antibody-based fusion proteins
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6242195B1 (en) 1998-04-02 2001-06-05 Genentech, Inc. Methods for determining binding of an analyte to a receptor
CA2323757C (en) 1998-04-02 2011-08-02 Genentech, Inc. Antibody variants and fragments thereof
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6284536B1 (en) 1998-04-20 2001-09-04 The Regents Of The University Of California Modified immunoglobin molecules and methods for use thereof
US7112324B1 (en) 1998-04-21 2006-09-26 Micromet Ag CD 19×CD3 specific polypeptides and uses thereof
US6455677B1 (en) 1998-04-30 2002-09-24 Boehringer Ingelheim International Gmbh FAPα-specific antibody with improved producibility
CA2331607A1 (en) 1998-05-06 1999-11-11 Temple University - Of The Commonwealth System Of Higher Education Reversal of proinflammatory response by ligating the macrophage fc.gamma.ri receptor
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
DE69942148D1 (en) * 1998-06-22 2010-04-29 Immunomedics Inc USE OF BISPECIFIC ANTIBODIES IN DIAGNOSIS AND THERAPY
GB9815909D0 (en) 1998-07-21 1998-09-16 Btg Int Ltd Antibody preparation
US20020142374A1 (en) 1998-08-17 2002-10-03 Michael Gallo Generation of modified molecules with increased serum half-lives
US6306926B1 (en) 1998-10-07 2001-10-23 3M Innovative Properties Company Radiopaque cationically polymerizable compositions comprising a radiopacifying filler, and method for polymerizing same
US7315786B2 (en) 1998-10-16 2008-01-01 Xencor Protein design automation for protein libraries
US6403312B1 (en) 1998-10-16 2002-06-11 Xencor Protein design automatic for protein libraries
US20020048772A1 (en) 2000-02-10 2002-04-25 Dahiyat Bassil I. Protein design automation for protein libraries
AU774334B2 (en) 1998-10-16 2004-06-24 Xencor Protein design automation for protein libraries
US20030049654A1 (en) 1998-10-16 2003-03-13 Xencor Protein design automation for protein libraries
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
KR100940380B1 (en) 1999-01-15 2010-02-02 제넨테크, 인크. Polypeptide Variants with Altered Effector Function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US6221556B1 (en) 1999-03-05 2001-04-24 General Electric Company Article for optical data storage device
US6723538B2 (en) 1999-03-11 2004-04-20 Micromet Ag Bispecific antibody and chemokine receptor constructs
EP2270148A3 (en) 1999-04-09 2011-06-08 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6939545B2 (en) 1999-04-28 2005-09-06 Genetics Institute, Llc Composition and method for treating inflammatory disorders
US6663862B1 (en) 1999-06-04 2003-12-16 Duke University Reagents for detection and purification of antibody fragments
JP2003507063A (en) 1999-08-20 2003-02-25 ジョンズ ホプキンス ユニバーシティ スクール オブ メディシン Methods and compositions for constructing and using fusion libraries
US6649165B2 (en) 1999-09-07 2003-11-18 Walter Schubert Method of blocking cytotoxic activity in patients with amyotrophic lateral sclerosis using soluble FcγRIII receptors
US7303749B1 (en) 1999-10-01 2007-12-04 Immunogen Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
CA2385528C (en) 1999-10-01 2013-12-10 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
EP1229125A4 (en) 1999-10-19 2005-06-01 Kyowa Hakko Kogyo Kk Process for producing polypeptide
ES2591102T3 (en) 1999-11-29 2016-11-24 The Trustees Of Columbia University In The City Of New York Isolation of five novel genes encoding new Fc receptor-type melanomas involved in the pathogenesis of lymphoma / melanoma
WO2001055319A2 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Endocrine related nucleic acids, proteins and antibodies
US20060272036A1 (en) 2000-02-03 2006-11-30 Lennart Hammarstrom Ruminant mhc class-i-like fc receptors
DE60113381T2 (en) 2000-02-10 2006-06-29 Xencor, Monrovia AUTOMATION IN PROTEIN DICE BY PROTEIN LIBRARIES
JP2003521931A (en) 2000-02-11 2003-07-22 バイオジェン インコーポレイテッド Heterologous polypeptides of the TNF family
WO2001062931A2 (en) 2000-02-25 2001-08-30 The Government Of The United States, As Represented By The Secretary Of The Department Of Health And Human Services ANTI-EGFRvIII SCFVS WITH IMPROVED CYTOTOXICITY AND YIELD, IMMUNOTOXINS BASED THEREON, AND METHODS OF USE THEREOF
US7687461B2 (en) 2000-03-02 2010-03-30 Xencor, Inc. Treatment of TNF-α related disorders with TNF-α variant proteins
US7056695B2 (en) 2000-03-02 2006-06-06 Xencor TNF-α variants
US7101974B2 (en) 2000-03-02 2006-09-05 Xencor TNF-αvariants
US7244823B2 (en) 2000-03-02 2007-07-17 Xencor TNF-alpha variants proteins for the treatment of TNF-alpha related disorders
US20010035606A1 (en) 2000-03-28 2001-11-01 Schoen Alan H. Set of blocks for packing a cube
JP2003531588A (en) 2000-04-11 2003-10-28 ジェネンテック・インコーポレーテッド Multivalent antibodies and their uses
JP2003531149A (en) 2000-04-13 2003-10-21 ザ・ロツクフエラー・ユニバーシテイ Enhancement of antibody-derived immune response
CA2407956A1 (en) 2000-05-03 2001-11-08 Amgen Inc. Modified peptides as therapeutic agents
US6358733B1 (en) 2000-05-19 2002-03-19 Apolife, Inc. Expression of heterologous multi-domain proteins in yeast
EP1278851B1 (en) 2000-05-19 2006-01-18 Scancell Limited Humanised antibodies to the epidermal growth factor receptor
AU2001264946A1 (en) 2000-05-24 2001-12-03 Imclone Systems Incorporated Bispecific immunoglobulin-like antigen binding proteins and method of production
CA2415902A1 (en) 2000-07-10 2002-01-17 Xencor Protein design automation for designing protein libraries with altered immunogenicity
AU2001283496A1 (en) * 2000-07-25 2002-02-05 Immunomedics, Inc. Multivalent target binding protein
US6333410B1 (en) 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
DE10043437A1 (en) 2000-09-04 2002-03-28 Horst Lindhofer Use of trifunctional bispecific and trispecific antibodies for the treatment of malignant ascites
US20030068649A1 (en) 2000-09-14 2003-04-10 Doberstein Stephen K. Methods and compositions for the construction and use of fusion libraries
AU2001294556A1 (en) 2000-09-14 2002-03-26 Xencor, Inc. Methods and compositions for the construction and use of fusion libraries
ES2620359T3 (en) 2000-10-06 2017-06-28 Kyowa Hakko Kirin Co., Ltd. Cells that produce antibody compositions
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
AU2001294175A1 (en) 2000-10-06 2002-04-22 Kyowa Hakko Kogyo Co. Ltd. Method of purifying antibody
US7064191B2 (en) 2000-10-06 2006-06-20 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
US7465790B2 (en) 2000-10-09 2008-12-16 Isis Innovation, Inc. Therapeutic antibodies
US20020128448A1 (en) 2000-10-20 2002-09-12 Idec Pharmaceuticals Corporation Variant IgG3 Rituxan and therapeutic use thereof
US6992234B2 (en) 2000-11-06 2006-01-31 The Jackson Laboratory FcRn-based therapeutics for the treatment of auto-immune disorders
US7235643B2 (en) 2000-11-07 2007-06-26 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with high affinity
GB0029407D0 (en) 2000-12-01 2001-01-17 Affitech As Product
US20040253242A1 (en) 2000-12-05 2004-12-16 Bowdish Katherine S. Rationally designed antibodies
AU2002248184C1 (en) 2000-12-12 2018-01-04 Board Of Regents, The University Of Texas System Molecules with extended half-lives, compositions and uses thereof
WO2002066653A2 (en) 2000-12-14 2002-08-29 Xencor Procaryotic libraries and uses
CA2430182C (en) 2000-12-14 2011-01-25 Genentech, Inc. Prokaryotically produced antibodies and uses thereof
US6979556B2 (en) 2000-12-14 2005-12-27 Genentech, Inc. Separate-cistron contructs for secretion of aglycosylated antibodies from prokaryotes
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
WO2002061093A1 (en) 2001-01-30 2002-08-08 Degussa Ag Nucleotide sequences which code for the otsa gene of c. glutamicum
AU2002251913A1 (en) 2001-02-02 2002-08-19 Millennium Pharmaceuticals, Inc. Hybrid antibodies and uses thereof
MXPA03007323A (en) 2001-02-19 2003-12-12 Merck Patent Gmbh Artificial proteins with reduced immunogenicity.
KR100899970B1 (en) 2001-02-19 2009-05-28 메르크 파텐트 게엠베하 Method for identification of t-cell epitopes and use for preparing molecules with reeduced immunogenicity
US20030049647A1 (en) 2001-02-22 2003-03-13 Bassil Dahiyat Use of nucleic acid libraries to create toxicological profiles
WO2002068453A2 (en) 2001-02-22 2002-09-06 Xencor Methods and compositions for the construction and use of fusion libraries using computational protein design methods
US20020168640A1 (en) 2001-02-22 2002-11-14 Min Li Biochips comprising nucleic acid/protein conjugates
CA2441595C (en) 2001-03-23 2012-07-03 Genencor International, Inc. Proteins producing an altered immunogenic response and methods of making and using the same
EP1243276A1 (en) 2001-03-23 2002-09-25 Franciscus Marinus Hendrikus De Groot Elongated and multiple spacers containing activatible prodrugs
US6992174B2 (en) 2001-03-30 2006-01-31 Emd Lexigen Research Center Corp. Reducing the immunogenicity of fusion proteins
AU2002307037B2 (en) 2001-04-02 2008-08-07 Biogen Idec Inc. Recombinant antibodies coexpressed with GnTIII
US7117096B2 (en) 2001-04-17 2006-10-03 Abmaxis, Inc. Structure-based selection and affinity maturation of antibody library
US6884869B2 (en) 2001-04-30 2005-04-26 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
KR20030033007A (en) 2001-05-31 2003-04-26 코울터 파머수티컬, 인코포레이티드 Cytotoxins, prodrugs, linkers and stabilizers useful therefor
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
BRPI0210405B8 (en) 2001-06-13 2021-05-25 Genmab As human monoclonal antibody, bispecific molecule, in vitro method to inhibit the growth of a cell expressing egfr, to induce cytolysis of a cell expressing egfr, and to detect the presence of egfr antigen or a cell expressing egfr in a sample, e, vector of expression
US20040242851A1 (en) 2001-06-26 2004-12-02 Zhenping Zhu Bispecific antibodies that bind to vegf receptors
CA2452824A1 (en) 2001-07-10 2003-01-23 Xencor Protein design automation for designing protein libraries with altered immunogenicity
GB0118662D0 (en) 2001-07-31 2001-09-19 Univ Southampton Binding agents
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
WO2003014325A2 (en) 2001-08-10 2003-02-20 Xencor Protein design automation for protein libraries
WO2003016470A2 (en) 2001-08-10 2003-02-27 University Of Virginia Patent Foundation Enhancing the efficacy of immunotherapies by supplementing with complement
KR100988949B1 (en) 2001-10-25 2010-10-20 제넨테크, 인크. Glycoprotein compositions
US6911321B2 (en) 2001-12-19 2005-06-28 Genentech, Inc. Non-human primate Fc receptors and methods of use
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
WO2003059282A2 (en) 2002-01-09 2003-07-24 Medarex, Inc. Human monoclonal antibodies against cd30
CA2472922A1 (en) 2002-02-11 2003-08-21 Genentech, Inc. Antibody variants with faster antigen association rates
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US20040110226A1 (en) 2002-03-01 2004-06-10 Xencor Antibody optimization
US20080219974A1 (en) 2002-03-01 2008-09-11 Bernett Matthew J Optimized antibodies that target hm1.24
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
JP2005526769A (en) 2002-03-15 2005-09-08 ザ・ブリガーム・アンド・ウーメンズ・ホスピタル・インコーポレーテッド Central airway administration for systemic delivery of therapeutic agents
EP1490085A2 (en) 2002-03-25 2004-12-29 Uab Research Foundation Fc receptor homolog, reagents, and uses thereof
US7332580B2 (en) 2002-04-05 2008-02-19 The Regents Of The University Of California Bispecific single chain Fv antibody molecules and methods of use thereof
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
JP2006513139A (en) 2002-07-03 2006-04-20 ザ・ブリガーム・アンド・ウーメンズ・ホスピタル・インコーポレーテッド Central airway administration for systemic delivery of therapeutic agents
JP2006500009A (en) 2002-07-09 2006-01-05 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
CA2494105C (en) 2002-07-31 2013-04-02 Seattle Genetics, Inc. Drug conjugates and their use for treating cancer, an autoimmune disease or an infectious disease
ES2381617T5 (en) 2002-08-14 2016-02-24 Macrogenics, Inc. Specific antibodies against FcgammaRIIB and its procedures for use
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
WO2004022717A2 (en) 2002-09-05 2004-03-18 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Minimally immunogenic variants of humanized col-1 antibody against carcinoembryonic antigen
EP1539811A4 (en) 2002-09-16 2006-05-24 Elusys Therapeutics Inc Production of bispecific molecules using polyethylene glycol linkers
EP2298805A3 (en) 2002-09-27 2011-04-13 Xencor, Inc. Optimized Fc variants and methods for their generation
US20060235208A1 (en) 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
US7820166B2 (en) 2002-10-11 2010-10-26 Micromet Ag Potent T cell modulating molecules
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7365168B2 (en) 2002-10-15 2008-04-29 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
SI1562972T1 (en) 2002-10-15 2010-12-31 Facet Biotech Corp Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
US7217797B2 (en) 2002-10-15 2007-05-15 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7705045B2 (en) 2002-11-14 2010-04-27 Syntarga, B.V. Prodrugs built as multiple self-elimination-release spacers
DK1572744T3 (en) 2002-12-16 2010-09-20 Genentech Inc Immunoglobulin variants and their applications
AU2004204942A1 (en) 2003-01-08 2004-07-29 Xencor, Inc Novel proteins with altered immunogenicity
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
CA2512729C (en) 2003-01-09 2014-09-16 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US7610156B2 (en) 2003-03-31 2009-10-27 Xencor, Inc. Methods for rational pegylation of proteins
EP1613649A2 (en) 2003-04-04 2006-01-11 Ingenium Pharmaceuticals AG Spinster-like protein genes, expression products, non-human animal model: uses in human metabolic disorders
PT2335725T (en) 2003-04-04 2017-01-06 Novartis Ag High concentration antibody and protein formulations
US20040219143A1 (en) * 2003-04-30 2004-11-04 Bluth Martin Heath Methods for recombinant immunoglobulin treatment
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
AR044388A1 (en) 2003-05-20 2005-09-07 Applied Molecular Evolution CD20 UNION MOLECULES
MX340862B (en) 2003-05-20 2016-07-28 Immunogen Inc Improved cytotoxic agents comprising new maytansinoids.
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
US20070041972A1 (en) 2003-05-30 2007-02-22 Alexion Pharmaceuticals, Inc. Antibodies and fusion proteins that include engineered constant regions
WO2004106381A1 (en) 2003-05-31 2004-12-09 Micromet Ag Pharmaceutical compositions comprising bispecific anti-cd3, anti-cd19 antibody constructs for the treatment of b-cell related disorders
RU2005137325A (en) 2003-05-31 2006-09-10 Микромет Аг (De) PHARMACEUTICAL COMPOSITION CONTAINING A DESIGN SPECIFIC TO ERS
US7888134B2 (en) 2003-06-05 2011-02-15 Oakland University Immunosensors: scFv-linker design for surface immobilization
DE602004011770T2 (en) 2003-06-12 2009-02-05 Eli Lilly And Co., Indianapolis FUSION PROTEIN
US20040254108A1 (en) 2003-06-13 2004-12-16 Jing Ma Preparation and application of anti-tumor bifunctional fusion proteins
NZ544923A (en) 2003-06-27 2009-02-28 Biogen Idec Inc Use of hydrophobic-interaction-chromatography or hinge-region modifications for the production of homogeneous anti-body solutions
US20050033029A1 (en) 2003-06-30 2005-02-10 Jin Lu Engineered anti-target immunoglobulin derived proteins, compositions, methods and uses
KR20060038461A (en) 2003-07-24 2006-05-03 이나뜨 파르마 Methods and compositions for increasing the efficiency of therapeutic antibodies using nk cell potentiating compounds
EP1653801A4 (en) 2003-07-26 2007-05-30 Biogen Idec Inc Altered antibodies having improved antigen-binding affinity
US20080050357A1 (en) 2003-08-01 2008-02-28 Claes Gustafsson Systems and Methods for Antibody Engineering
NZ545776A (en) 2003-08-22 2009-05-31 Biogen Idec Inc Improved antibodies having altered effector function and methods for making the same
US20050152894A1 (en) 2003-09-05 2005-07-14 Genentech, Inc. Antibodies with altered effector functions
CA2579635A1 (en) 2003-09-10 2005-03-17 Baxter International Inc. Peptides that inhibit complement activation
US20060134105A1 (en) 2004-10-21 2006-06-22 Xencor, Inc. IgG immunoglobulin variants with optimized effector function
US20150071948A1 (en) 2003-09-26 2015-03-12 Gregory Alan Lazar Novel immunoglobulin variants
WO2005037867A1 (en) 2003-10-15 2005-04-28 Pdl Biopharma, Inc. ALTERATION OF Fc-FUSION PROTEIN SERUM HALF-LIVES BY MUTAGENESIS OF POSITIONS 250, 314 AND/OR 428 OF THE HEAVY CHAIN CONSTANT REGION OF IG
US20050176028A1 (en) 2003-10-16 2005-08-11 Robert Hofmeister Deimmunized binding molecules to CD3
GB0324368D0 (en) 2003-10-17 2003-11-19 Univ Cambridge Tech Polypeptides including modified constant regions
ZA200603619B (en) 2003-11-06 2008-10-29 Seattle Genetics Inc Monomethylvaline compounds capable of conjugation to ligands
CA2545603A1 (en) 2003-11-12 2005-05-26 Biogen Idec Ma Inc. Neonatal fc receptor (fcrn)-binding polypeptide variants, dimeric fc binding proteins and methods related thereto
WO2005063815A2 (en) 2003-11-12 2005-07-14 Biogen Idec Ma Inc. Fcϝ receptor-binding polypeptide variants and methods related thereto
US20050142133A1 (en) 2003-12-03 2005-06-30 Xencor, Inc. Optimized proteins that target the epidermal growth factor receptor
AU2004297616B2 (en) 2003-12-04 2008-12-18 Xencor, Inc. Methods of generating variant proteins with increased host string content and compositions thereof
ES2384569T3 (en) 2003-12-19 2012-07-09 Genentech, Inc. Fragments of monovalent antibodies useful as therapeutic agents
US7235641B2 (en) 2003-12-22 2007-06-26 Micromet Ag Bispecific antibodies
ATE437184T1 (en) 2004-01-12 2009-08-15 Applied Molecular Evolution VARIANTS OF THE FC REGION
CA2561264A1 (en) 2004-03-24 2005-10-06 Xencor, Inc. Immunoglobulin variants outside the fc region
WO2005123780A2 (en) 2004-04-09 2005-12-29 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
MXPA06011796A (en) 2004-04-16 2007-05-07 Macrogenics Inc FCgammaRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF.
MXPA06011805A (en) 2004-04-16 2006-12-15 Genentech Inc Method for augmenting b cell depletion.
KR101297146B1 (en) 2004-05-10 2013-08-21 마크로제닉스, 인크. HUMANIZED FcγRIIB SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
AU2005244980B2 (en) 2004-05-19 2011-09-15 E. R. Squibb & Sons, L.L.C. Chemical linkers and conjugates thereof
US7691962B2 (en) 2004-05-19 2010-04-06 Medarex, Inc. Chemical linkers and conjugates thereof
KR100545720B1 (en) 2004-05-31 2006-01-24 메덱스젠 주식회사 Glycosylated Immunoglobulin and Immunoadhesin comprising the same
EP1753783B1 (en) 2004-06-03 2014-08-06 Novimmune SA Anti-cd3 antibodies and methods of use thereof
US20060018897A1 (en) 2004-06-28 2006-01-26 Transtarget Inc. Bispecific antibodies
WO2006085967A2 (en) 2004-07-09 2006-08-17 Xencor, Inc. OPTIMIZED ANTI-CD20 MONOCONAL ANTIBODIES HAVING Fc VARIANTS
ES2530340T3 (en) 2004-07-15 2015-03-02 Xencor Inc Optimized Fc variants
WO2006020258A2 (en) 2004-07-17 2006-02-23 Imclone Systems Incorporated Novel tetravalent bispecific antibody
ZA200700947B (en) 2004-07-23 2008-06-25 Genentech Inc Crystallization of antibodies or fragments thereof
ES2426816T3 (en) 2004-08-04 2013-10-25 Mentrik Biotech, Llc Fc regions variants
ZA200701715B (en) 2004-08-19 2008-07-30 Genentech Inc Polypeptide variants with altered effector function
RU2404991C2 (en) 2004-09-02 2010-11-27 Дженентек, Инк. FcγRIIB RECEPTOR ANTIBODIES AND APPLICATION THEREOF
KR101270829B1 (en) 2004-09-23 2013-06-07 제넨테크, 인크. Cystein engineered antibodies and conjugates
BRPI0516011A (en) 2004-09-24 2008-08-19 Amgen Inc modified fc molecules
EP1810035A4 (en) 2004-11-10 2010-03-17 Macrogenics Inc Engineering fc antibody regions to confer effector function
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US20070135620A1 (en) 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
US9200079B2 (en) 2004-11-12 2015-12-01 Xencor, Inc. Fc variants with altered binding to FcRn
US8066989B2 (en) 2004-11-30 2011-11-29 Trion Pharma Gmbh Method of treating tumor growth and metastasis by using trifunctional antibodies to reduce the risk for GvHD in allogeneic antitumor cell therapy
EP1772465B1 (en) 2005-01-05 2009-02-18 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Synthetic immunoglobulin domains with binding properties engineered in regions of the molecule different from the complementarity determining regions
CA2595169A1 (en) 2005-01-12 2006-07-20 Xencor, Inc. Antibodies and fc fusion proteins with altered immunogenicity
US8716451B2 (en) 2005-01-12 2014-05-06 Kyowa Hakko Kirin Co., Ltd Stabilized human IgG2 and IgG3 antibodies
US20110123440A1 (en) 2005-03-29 2011-05-26 Genevieve Hansen Altered Antibody FC Regions and Uses Thereof
WO2006105338A2 (en) 2005-03-31 2006-10-05 Xencor, Inc. Fc VARIANTS WITH OPTIMIZED PROPERTIES
EP3050963B1 (en) 2005-03-31 2019-09-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
US7714016B2 (en) 2005-04-08 2010-05-11 Medarex, Inc. Cytotoxic compounds and conjugates with cleavable substrates
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2006130834A2 (en) 2005-05-31 2006-12-07 Board Of Regents, The University Of Texas System IGGl ANTIBODIES WITH MUTATED FC PORTION FOR INCREASED BINDING TO FCRN RECEPTOR AND USES THEREOF
EP1899477A4 (en) 2005-07-01 2010-01-20 Medimmune Inc An integrated approach for generating multidomain protein therapeutics
WO2007015168A2 (en) 2005-07-01 2007-02-08 Dako Denmark A/S Monomeric and polymeric linkers useful for conjugating biological molecules and other substances
CA2652434A1 (en) 2005-07-08 2007-01-18 Xencor, Inc. Optimized proteins that target ep-cam
KR20080047540A (en) 2005-07-25 2008-05-29 트루비온 파마슈티칼스, 인코포레이티드 Single dose use of cd20-specific binding molecules
PL2298815T3 (en) 2005-07-25 2015-08-31 Emergent Product Dev Seattle B-cell reduction using CD37-specific and CD20-specific binding molecules
JP2009509918A (en) 2005-08-05 2009-03-12 シンタルガ・ビーブイ Triazole-containing releasable linkers, their conjugates, and production methods
CA2618681C (en) 2005-08-10 2015-10-27 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
AU2006299429B2 (en) 2005-10-03 2012-02-23 Xencor, Inc. Fc variants with optimized Fc receptor binding properties
EP1951757B1 (en) 2005-10-06 2014-05-14 Xencor, Inc. Optimized anti-cd30 antibodies
CA2625440C (en) 2005-10-11 2023-06-13 Micromet Ag Compositions comprising cross-species-specific antibodies and uses thereof
CA2625619A1 (en) 2005-10-14 2007-04-26 Medimmune, Inc. Cell display of antibody libraries
US20070087005A1 (en) 2005-10-14 2007-04-19 Lazar Gregory A Anti-glypican-3 antibody
WO2007047829A2 (en) 2005-10-19 2007-04-26 Laboratoires Serono S.A. Novel heterodimeric proteins and uses thereof
AR056142A1 (en) 2005-10-21 2007-09-19 Amgen Inc METHODS TO GENERATE THE MONOVALENT IGG ANTIBODY
WO2007059404A2 (en) 2005-11-10 2007-05-24 Medarex, Inc. Duocarmycin derivatives as novel cytotoxic compounds and conjugates
AU2006318580A1 (en) 2005-11-21 2007-05-31 Merck Serono Sa Compositions and methods of producing hybrid antigen binding molecules and uses thereof
CA2633713A1 (en) 2005-12-21 2007-06-28 Micromet Ag Epha2 bite molecules and uses thereof
BRPI0707426A2 (en) 2006-02-02 2011-05-03 Syntarga Bv compound, conjugate, use of a compound, pharmaceutical composition, process for preparing a pharmaceutical composition, and methods of treating a mammal in need thereof, and treating or preventing a tumor in a mammal
WO2007092772A2 (en) 2006-02-03 2007-08-16 Medimmune, Inc. Protein formulations
EP1820513A1 (en) 2006-02-15 2007-08-22 Trion Pharma Gmbh Destruction of tumor cells expressing low to medium levels of tumor associated target antigens by trifunctional bispecific antibodies
EP1829895A1 (en) 2006-03-03 2007-09-05 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Bispecific molecule binding TLR9 and CD32 and comprising a T cell epitope for treatment of allergies
GEP20135917B (en) 2006-03-17 2013-09-10 Biogen Idec Inc Stabilized polypeptide compositions
ES2395969T3 (en) 2006-03-24 2013-02-18 Merck Patent Gmbh Genetically modified heterodimeric protein domains
WO2007114319A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Method for control of blood kinetics of antibody
CN105177091A (en) 2006-03-31 2015-12-23 中外制药株式会社 Antibody modification method for purifying bispecific antibody
JP2009541275A (en) 2006-06-22 2009-11-26 ノボ・ノルデイスク・エー/エス Production of bispecific antibodies
HUE030269T2 (en) 2006-06-26 2017-04-28 Macrogenics Inc Fc riib-specific antibodies and methods of use thereof
AT503902B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING IMMUNE LOBULINS
AT503861B1 (en) 2006-07-05 2008-06-15 F Star Biotech Forsch & Entw METHOD FOR MANIPULATING T-CELL RECEPTORS
AT503889B1 (en) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F MULTIVALENT IMMUNE LOBULINE
US20100166741A1 (en) 2006-07-13 2010-07-01 Genentech , Inc. Altered br-3 binding polypeptides
DK2069558T3 (en) 2006-10-02 2013-08-05 Sea Lane Biotechnologies Llc Design and construction of diverse synthetic peptide and polypeptide libraries
US20080112961A1 (en) 2006-10-09 2008-05-15 Macrogenics, Inc. Identification and Engineering of Antibodies with Variant Fc Regions and Methods of Using Same
JP2010506842A (en) 2006-10-16 2010-03-04 メディミューン,エルエルシー Molecules with reduced half-life, compositions thereof and uses
GB0620934D0 (en) 2006-10-20 2006-11-29 Cambridge Antibody Tech Protein variants
WO2008070780A1 (en) 2006-12-07 2008-06-12 Novartis Ag Antagonist antibodies against ephb3
US8652466B2 (en) 2006-12-08 2014-02-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
AR064464A1 (en) 2006-12-22 2009-04-01 Genentech Inc ANTIBODIES ANTI - INSULINAL GROWTH FACTOR RECEIVER
CN101679974B (en) 2007-03-27 2015-09-30 航道生物技术有限责任公司 Comprise construct and the library of antibody surrogate light chain sequences
EP1975178A1 (en) 2007-03-30 2008-10-01 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Transcytotic modular antibody
KR101589759B1 (en) 2007-04-03 2016-01-29 암젠 리서치 (뮌헨) 게엠베하 Cross-species-specific cd3-epsilon binding domain
MX2009010611A (en) 2007-04-03 2010-03-26 Micromet Ag Cross-species-specific bispecific binders.
WO2008124858A2 (en) 2007-04-11 2008-10-23 F-Star Biotechnologische Forschungs- Und Entwicklungsges. M.B.H. Targeted receptor
EP2599791A1 (en) 2007-04-27 2013-06-05 Genentech, Inc. Potent, stable and non-immunosuppressive anti-CD4 antibodies
AU2008260498B2 (en) 2007-05-30 2012-11-29 Xencor, Inc. Methods and compositions for inhibiting CD32b expressing cells
WO2008145142A1 (en) 2007-05-31 2008-12-04 Genmab A/S Stable igg4 antibodies
BRPI0812562A2 (en) 2007-06-12 2019-09-24 Trubion Pharmaceuticals Inc anti-cd20 therapeutic compositions and methods
CA2688667A1 (en) 2007-06-26 2008-12-31 Medimmune, Llc Methods of treating rsv infections and related conditions
JP5602625B2 (en) 2007-06-26 2014-10-08 エフ−スター ビオテヒノロギッシェ フォルシュングス− ウント エントヴィッケルングスゲゼルシャフト ミット ベシュレンクテル ハフツング Binding substance display
AU2008282729B2 (en) 2007-08-01 2015-07-16 Scott & White Healthcare A fold-back diabody diphtheria toxin immunotoxin and methods of use
US8680293B2 (en) 2007-08-01 2014-03-25 Syntarga B.V. Substituted CC-1065 analogs and their conjugates
JP2010538012A (en) 2007-08-28 2010-12-09 バイオジェン アイデック マサチューセッツ インコーポレイテッド Compositions that bind to multiple epitopes of IGF-1R
EP2033657A1 (en) 2007-09-04 2009-03-11 Trion Pharma Gmbh Intraoperative trifunctional antibody application for prophylatic intraperitonal tumour cell dissemination
US20100226925A1 (en) 2007-09-14 2010-09-09 Amgen Inc. Homogeneous Antibody Populations
EP2535351A3 (en) 2007-09-26 2013-04-03 UCB Pharma S.A. Dual specificity antibody fusions
EP3789400A1 (en) 2007-09-26 2021-03-10 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
EP2202245B1 (en) 2007-09-26 2016-08-24 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
JP5298134B2 (en) 2007-11-01 2013-09-25 パーシード セラピューティクス リミテッド ライアビリティ カンパニー Immunosuppressive polypeptides and nucleic acids
EP3825329A1 (en) 2007-12-26 2021-05-26 Xencor, Inc. Fc variants with altered binding to fcrn
PT2235064E (en) 2008-01-07 2016-03-01 Amgen Inc Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
WO2009106096A1 (en) 2008-02-27 2009-09-03 Fresenius Biotech Gmbh Treatment of resistant tumors with trifunctional antibodies
NZ621443A (en) 2008-04-11 2015-09-25 Emergent Product Dev Seattle Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
CN102076865B (en) 2008-05-02 2016-03-16 西雅图基因公司 The antibody reduced for the manufacture of core fucosylation and the method and composition of antibody derivatives
EP2297209A4 (en) 2008-06-03 2012-08-01 Abbott Lab Dual variable domain immunoglobulins and uses thereof
US20170247470A9 (en) 2008-09-17 2017-08-31 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
WO2010033736A1 (en) 2008-09-17 2010-03-25 Xencor, Inc. Novel compositons and methods for treating ige-mediated disorders
HRP20160857T4 (en) 2008-10-01 2022-12-09 Amgen Research (Munich) Gmbh Bispecific single chain antibodies with specificity for high molecular weight target antigens
BRPI0919840B1 (en) 2008-10-01 2023-02-28 Amgen Research (Munich) Gmbh BISPECIFIC SINGLE CHAIN ANTIBODY MOLECULE, ITS USE, AND PHARMACEUTICAL COMPOSITION COMPRISING IT
AU2009299794B2 (en) 2008-10-01 2015-08-13 Amgen Research (Munich) Gmbh Cross-species-specific single domain bispecific single chain antibody
DK2356153T3 (en) 2008-10-01 2016-07-04 Amgen Res (Munich) Gmbh Bispecific single CHAIN ​​PSMAXCD3 ANTIBODY THAT ARE SPECIFICALLY TO ALL SPECIES
MX2011003763A (en) 2008-10-10 2011-04-27 Emergent Product Dev Seattle Tcr complex immunotherapeutics.
JP5677970B2 (en) 2008-11-03 2015-02-25 シンタルガ・ビーブイ Novel CC-1065 analogs and complexes thereof
JP2012515556A (en) 2009-01-23 2012-07-12 バイオジェン・アイデック・エムエイ・インコーポレイテッド Stabilized Fc polypeptides with reduced effector function and methods of use
EP2233500A1 (en) 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
EP2241576A1 (en) 2009-04-17 2010-10-20 Trion Pharma Gmbh Use of trifunctional bispecific antibodies for the treatment of tumors associated with CD133+/EpCAM+ cancer stem cells
EP2975051B1 (en) 2009-06-26 2021-04-14 Regeneron Pharmaceuticals, Inc. Readily isolated bispecific antibodies with native immunoglobulin format
WO2010151808A1 (en) 2009-06-26 2010-12-29 Sea Lane Biotechnologies, Llc Expression of surrogate light chains
US9308258B2 (en) 2009-07-08 2016-04-12 Amgen Inc. Stable and aggregation free antibody FC molecules through CH3 domain interface engineering
JPWO2011013746A1 (en) 2009-07-31 2013-01-10 株式会社アルバック Deposition equipment
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
DE102009045006A1 (en) 2009-09-25 2011-04-14 Technische Universität Dresden Anti-CD33 antibodies and their use for immuno-targeting in the treatment of CD33-associated diseases
CA2781539C (en) 2009-11-23 2021-07-20 Amgen Inc. Monomeric antibody fc
US20130017199A1 (en) 2009-11-24 2013-01-17 AMPLIMMUNE ,Inc. a corporation Simultaneous inhibition of pd-l1/pd-l2
KR101791430B1 (en) 2009-11-30 2017-10-30 얀센 바이오테크 인코포레이티드 ANTIBODY Fc MUTANTS WITH ABLATED EFFECTOR FUNCTIONS
KR102103093B1 (en) 2009-12-25 2020-04-21 추가이 세이야쿠 가부시키가이샤 Polypeptide modification method for purifying polypeptide multimers
US20130129723A1 (en) 2009-12-29 2013-05-23 Emergent Product Development Seattle, Llc Heterodimer Binding Proteins and Uses Thereof
HUE029257T2 (en) 2009-12-29 2017-02-28 Aptevo Res And Dev Llc Heterodimer binding proteins and uses thereof
US20110189178A1 (en) 2010-02-04 2011-08-04 Xencor, Inc. Immunoprotection of Therapeutic Moieties Using Enhanced Fc Regions
US8153437B2 (en) 2010-03-10 2012-04-10 National Center For Biological Sciences DNA-based molecular switches and uses thereof
EP2552957A4 (en) 2010-03-29 2013-11-20 Zymeworks Inc Antibodies with enhanced or suppressed effector function
CA2796181C (en) 2010-04-20 2023-01-03 Genmab A/S Heterodimeric antibody fc-containing proteins and methods for production thereof
TWI586806B (en) 2010-04-23 2017-06-11 建南德克公司 Production of heteromultimeric proteins
US9527926B2 (en) 2010-05-14 2016-12-27 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
SI2580243T1 (en) 2010-06-09 2020-02-28 Genmab A/S Antibodies against human cd38
EP3029066B1 (en) 2010-07-29 2019-02-20 Xencor, Inc. Antibodies with modified isoelectric points
JP5964300B2 (en) 2010-08-02 2016-08-03 マクロジェニクス,インコーポレーテッド Covalently bonded diabody and its use
WO2012032080A1 (en) 2010-09-07 2012-03-15 F-Star Biotechnologische Forschungs- Und Entwicklungsges.M.B.H Stabilised human fc
EP2635607B1 (en) 2010-11-05 2019-09-04 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
US9266938B2 (en) 2011-02-10 2016-02-23 Roche Glycart Ag Mutant interleukin-2 polypeptides
CN103732628B (en) 2011-03-03 2017-06-23 酵活有限公司 Multivalence heteropolymer frame design and construct
MX2013010172A (en) 2011-03-11 2013-10-25 Amgen Inc Method of correlated mutational analysis to improve therapeutic antibodies.
US20140112926A1 (en) 2011-03-16 2014-04-24 Amgen Inc. Fc VARIANTS
EA201791693A1 (en) 2011-03-25 2018-05-31 Гленмарк Фармасьютикалс С.А. HETERODIMERNY IMMUNOHLOBULINS
TWI588156B (en) 2011-03-28 2017-06-21 賽諾菲公司 Dual variable region antibody-like binding proteins having cross-over binding region orientation
MX336740B (en) 2011-03-29 2016-01-29 Roche Glycart Ag Antibody fc variants.
EA201892619A1 (en) 2011-04-29 2019-04-30 Роше Гликарт Аг IMMUNOCONJUGATES CONTAINING INTERLEUKIN-2 MUTANT POLYPETIPS
ES2732213T3 (en) 2011-05-21 2019-11-21 Macrogenics Inc Molecules that bind to CD3 capable of binding to human and non-human CD3
KR20140028013A (en) 2011-05-25 2014-03-07 머크 샤프 앤드 돔 코포레이션 Method for preparing fc-containing polypeptides having improved properties
EP2729488A4 (en) 2011-07-06 2015-01-14 Medimmune Llc Methods for making multimeric polypeptides
WO2013016714A1 (en) 2011-07-28 2013-01-31 Sea Lane Biotechnologies Sur-binding proteins against erbb3
WO2013022855A1 (en) 2011-08-05 2013-02-14 Xencor, Inc. Antibodies with modified isoelectric points and immunofiltering
EA030147B1 (en) 2011-08-23 2018-06-29 Роше Гликарт Аг Bispecific t cell activating antigen binding molecules
CA2843158A1 (en) 2011-08-26 2013-03-07 Merrimack Pharmaceuticals, Inc. Tandem fc bispecific antibodies
EP3939996A1 (en) 2011-09-30 2022-01-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
WO2013055809A1 (en) 2011-10-10 2013-04-18 Xencor, Inc. A method for purifying antibodies
RU2018112861A (en) 2011-10-20 2019-03-04 ИЭсБиЭйТЕК - Э НОВАРТИС КОМПАНИ ЭлЭлСи STABLE ANTIBODIES COMBINING WITH MULTIPLE ANTIGENS
CA2853230C (en) 2011-10-31 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule having regulated conjugation between heavy-chain and light-chain
MX358862B (en) 2011-11-04 2018-09-06 Zymeworks Inc Stable heterodimeric antibody design with mutations in the fc domain.
ES2710916T3 (en) 2011-12-22 2019-04-29 I2 Pharmaceuticals Inc Substitute binding proteins
AU2013223087B2 (en) 2012-02-24 2018-02-22 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting disappearance of antigen via FcgammaRIIb
PL2838918T3 (en) 2012-04-20 2019-11-29 Merus Nv Methods and means for the production of heterodimeric ig-like molecules
DK2857419T3 (en) 2012-05-30 2021-03-29 Chugai Pharmaceutical Co Ltd Antigen-binding molecule for the elimination of aggregated antigens
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
KR20150036606A (en) 2012-07-13 2015-04-07 자임워크스 인코포레이티드 Bispecific asymmetric heterodimers comprising anti-cd3 constructs
CN104640561A (en) 2012-07-23 2015-05-20 酵活有限公司 Immunoglobulin constructs comprising selective pairing of the light and heavy chains
US20140377269A1 (en) 2012-12-19 2014-12-25 Adimab, Llc Multivalent antibody analogs, and methods of their preparation and use
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
LT2943511T (en) 2013-01-14 2019-11-11 Xencor Inc Novel heterodimeric proteins
EP2945969A1 (en) 2013-01-15 2015-11-25 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US20140294812A1 (en) 2013-03-15 2014-10-02 Xencor, Inc. Fc variants that improve fcrn binding and/or increase antibody half-life
WO2014145907A1 (en) 2013-03-15 2014-09-18 Xencor, Inc. Targeting t cells with heterodimeric proteins
EP3066133A1 (en) 2013-11-04 2016-09-14 Glenmark Pharmaceuticals S.A. Production of t cell retargeting hetero-dimeric immunoglobulins
KR102327996B1 (en) 2014-03-28 2021-11-17 젠코어 인코포레이티드 Bispecific antibodies that bind to cd38 and cd3
US20160060360A1 (en) 2014-07-24 2016-03-03 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
WO1992011018A1 (en) 1990-12-19 1992-07-09 Protein Design Labs, Inc. Improved humanized immunoglobulins
WO1995009917A1 (en) * 1993-10-07 1995-04-13 The Regents Of The University Of California Genetically engineered bispecific tetravalent antibodies
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US20060134150A1 (en) 2000-12-22 2006-06-22 Jane Werling Submicron suspensions with polymorph control
WO2010028796A1 (en) * 2008-09-10 2010-03-18 F. Hoffmann-La Roche Ag Trispecific hexavalent antibodies
WO2010034441A1 (en) * 2008-09-26 2010-04-01 F. Hoffmann-La Roche Ag Bispecific anti-egfr/anti-igf-1r antibodies
WO2010112193A1 (en) * 2009-04-02 2010-10-07 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
WO2010115552A1 (en) * 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-3/anti-c-met antibodies
WO2010115589A1 (en) * 2009-04-07 2010-10-14 Roche Glycart Ag Trivalent, bispecific antibodies
WO2010115551A1 (en) * 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-1/anti-c-met antibodies
WO2010115553A1 (en) * 2009-04-07 2010-10-14 Roche Glycart Ag Bispecific anti-erbb-2/anti-c-met antibodies

Non-Patent Citations (99)

* Cited by examiner, † Cited by third party
Title
"A method for making multispecific antibodies having heteromultimeric and common components", EXPERT OPINION ON THERAPEUTIC PATENTS 1999 GB, vol. 9, no. 6, 1999, pages 785 - 790, XP002610994, ISSN: 1354-3776 *
"Molecular Cloning - A Laboratory Manual,3rd Ed.", 2001, COLD SPRING HARBOR LABORATORY PRESS
"WHO Review of the notation for the allotypic and related markers of human immunoglobulins", EUR. J. IMMUNOL., vol. 6, 1976, pages 599 - 601
"WHO Review of the notation for the allotypic and related markers of human immunoglobulins", J IMMUNOGEN, vol. 3, 1976, pages 357 - 362
ALLEY ET AL., CURR OPIN CHEM BIOL, vol. 14, no. 4, 2010, pages 529 - 37
ASHKENAZI ET AL., CURR OPIN IMMUNOL, vol. 9, 1997, pages 195 - 200
BAEUERLE; REINHARDT, CANCER RES, vol. 69, no. 12, 2009, pages 4941 - 4
BOWLES; WEINER, J IMMUNOL METHODS, vol. 304, 2005, pages 88 - 99
CARTER ET AL., PROC NATL ACAD SCI USA, vol. 89, 1992, pages 4285 - 4289
CARTER, NATURE REVIEWS IMMUNOLOGY, vol. 6, 2006, pages 343 - 357
CHAMES; BATY, MABS, vol. 1, no. 6, 2009, pages 1 - 9
CHAMOW ET AL., TRENDS BIOTECHNOL, vol. 14, 1996, pages 52 - 60
CHAN; CARTER, NATURE REVIEWS IMMUNOLOGY, vol. 10, 2010, pages 301 - 316
CHATENOUD; BLUESTONE, NATURE REVIEWS IMMUNOLOGY, vol. 7, 2007, pages 622 - 632
CLARK, IGG EFFECTOR MECHANISMS, CHEM IMMUNOL., vol. 65, 1997, pages 88 - 110
COX ET AL., NAT BIOTECHNOL, vol. 24, no. 12, 2006, pages 1591 - 7
DALL ACQUA ET AL., JOURNAL OF IMMUNOLOGY, vol. 169, 2002, pages 5171 - 5180
DALL'ACQUA ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 281, 2006, pages 23514 - 23524
DAVIES ET AL., BIOTECHNOL BIOENG, vol. 74, 2001, pages 288 - 294
DAVIS ET AL., PROTEIN ENGINEERING, DESIGN & SELECTION, vol. 23, no. 4, 2010, pages 195 - 202
DUROCHER ET AL., NUCLEIC ACIDS RESEARCH, vol. 30, 2002, pages E9
EDELMAN ET AL., PROC NATL ACAD SCI USA, vol. 63, 1969, pages 78 - 85
FISCHER NICOLAS ET AL: "Bispecific antibodies: Molecules that enable novel therapeutic strategies", PATHOBIOLOGY, KARGER, BASEL, CH, vol. 74, no. 1, 1 May 2007 (2007-05-01), pages 3 - 14, XP008102855, ISSN: 1015-2008, DOI: DOI:10.1159/000101046 *
GHETIE ET AL., ANNU REV IMMUNOL, vol. 18, 2000, pages 739 - 766
GHETIE ET AL., ANNU REV TMMUNOI, vol. 18, 2000, pages 739 - 766
GORMAN; CLARK, SEMIN IMMUNOL, vol. 2, no. 6, 1990, pages 457 - 66
GUNASEKARAN ET AL., J BIOL CHEM, vol. 285, no. 25, 2010, pages 1937 - 19646
HAYHURST; GEORGIOU, CURR OPIN CHEM BIOL, vol. 5, 2001, pages 683 - 689
HEYMAN, IMMUNOL LETT, vol. 88, no. 2, 2003, pages 157 - 161
HEYMAN, IMMUNOL LETT, vol. 88, no. 21, 2003, pages 157 - 161
HINTON ET AL., J. BIOL. CHEM., vol. 279, no. 8, 2004, pages 6213 - 6216
HINTON ET AL., JOURNAL OF IMMUNOLOGY, vol. 176, 2006, pages 346 - 356
HOLLIGER, P. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOLLIGER; HUDSON, NATURE BIOTECHNOLOGY, vol. 23, no. 9, 2005, pages 1126 - 1136
HOLLIGER; HUDSON, NATURE BIOTECHNOLOGY, vol. 23, no. 9, 2006, pages 1126 - 1136
HUANG, CURR OPIN BIOTECHNOLOGY, vol. 20, 2009, pages 692 - 699
HUBBARD ET AL., ANN. REV. BIOCHEM., vol. 50, 1981, pages 555 - 583
JEFFERIS ET AL., IMMUNOL LETT, vol. 82, 2002, pages 57 - 65
JONES, NATURE, vol. 321, 1986, pages 522 - 525
KABAT ET AL.: "Sequences of Proteins of Immunological interest", 1991, UNITED STATES PUBLIC HEALTH SERVICE
KABAT: "Sequences of Proteins of Immunological Interest, 5th Ed.,", 1991, UNITED STATES PUBLIC HEALTH SERVICE
KANEKO ET AL., SCIENCE, vol. 313, 2006, pages 670 - 673
KIM ET AL., J. MOL. EVOL., vol. 54, 2001, pages 1 - 9
KJER-NIELSEN ET AL., PROC NATL ACAD SCI USA, vol. 101, 2004, pages 7675 - 7680
KONTERMANN , DUBEL,: "Antibody Engineering", 2001, SPRINGER
KORTT A A ET AL: "Dimeric and trimeric antibodies: high avidity scFvs for cancer targeting", BIOMOLECULAR ENGINEERING, ELSEVIER, NEW YORK, NY, US, vol. 18, no. 3, 15 October 2001 (2001-10-15), pages 95 - 108, XP004305907, ISSN: 1389-0344, DOI: DOI:10.1016/S1389-0344(01)00090-9 *
KUHNS ET AL., IMMUNITY, vol. 24, 2006, pages 133 - 139
LAZAR ET AL., MOL IMMUNOL, vol. 44, 2007, pages 1986 - 1998
LEFRANC ET AL.: "The human IgG subclasses: molecular analysis of structure, function and regulation", 1990, PERGAMON, pages: 43 - 78
LEFRANC, G. ET AL., HUM. GENET., vol. 50, 1979, pages 199 - 211
LI ET AL., NATURE BIOTECHNOLOGY, vol. 24, no. 2, 2006, pages 210 - 215
LIU; SCHULTZ, ANNU REV BIOCHEM, vol. 79, 2010, pages 413 - 444
LOGHEM E VAN: "Allotypic markers", MONOGR ALLERGY, vol. 19, 1986, pages 40 - 51
LONBERG, CURR OPIN IMMUNOL., vol. 20, no. 4, 2008, pages 450 - 9
LONBERG, HANDB EXP PHARMACOL, vol. 181, 2008, pages 69 - 97
LU ET AL., J BIOL CHEM, vol. 280, no. 20, 2005, pages 19665 - 19672
MARQUEZ ET AL., CELL LMMUNOL, vol. 264, no. 1, 2010, pages 86 - 92
MAYNARD ET AL., ANNU REV BIOMED ENG, vol. 2, 2000, pages 339 - 376
MAYNARD; GEORGIOU, ANNU REV BIOMED ENG, vol. 2, 2000, pages 339 - 76
MECHETINA ET AL., IMMUNOGENETICS, vol. 54, 2002, pages 463 - 468
MICHAELSON ET AL., MABS, vol. 1, no. 2, 2009, pages 128 - 141
MILSTEIN ET AL., NATURE, vol. 305, 1983, pages 537 - 540
MONDON ET AL., FRONT BIOSCI, vol. 13, 2008, pages 1117 - 29
MOREA ET AL., BIOPHYS CHEM, vol. 68, 1997, pages 9 - 16
MOREA ET AL., METHODS, vol. 20, 2000, pages 267 - 279
NECHANSKY ET AL., MOL IMMUNJOL, vol. 44, no. 7, 2007, pages 1826 - 8
OSOL, A.: "Remington's Pharmaceutical Sciences 16th edition,", 1980
PENICHET ET AL., J. LMMUNOL. METHODS, vol. 248, 2001, pages 91 - 101
PERRUCHE ET AL., J IMMUNOL, vol. 183, no. 2, 2009, pages 953 - 61
POLJAK, R. J. ET AL., STRUCTURE, vol. 2, 1994, pages 1121 - 1123
RAGHAVAN ET AL., ANNU REV CELL DEV BIOL, vol. 12, 1996, pages 181 - 220
RAVETCH ET AL., ANNU REV IMMUNOL, vol. 19, 2001, pages 275 - 290
REICHERT ET AL., NATURE BIOTECHNOLOGY, vol. 23, no. 9, 2005, pages 1073 - 1078
RICHARDS ET AL., MOL CANCER THER, vol. 7, no. 8, 2008, pages 2517 - 27
RICHARDS ET AL., MOL CANCER THER, vol. 7, no. 81, 2008, pages 2517 - 27
RIDGEWAY ET AL., PROTEIN ENGINEERING, vol. 9, no. 71, 1996, pages 617 - 621
RIDGWAY J B ET AL: "'Knobs-into-holes' engineering of antibody CH3 domains for heavy chain heterodimerization.", PROTEIN ENGINEERING JUL 1996 LNKD- PUBMED:8844834, vol. 9, no. 7, July 1996 (1996-07-01), pages 617 - 621, XP002610995, ISSN: 0269-2139 *
SCALLON ET AL., MOL IMMUNOL., vol. 44, no. 7, 2007, pages 1524 - 34
SCOPES: "Protein Purification: Principles and Practice", 1994, SPRINGER-VERLAG
SHEN ET AL., J BIOL CHEM, vol. 281, no. 16, 2006, pages 10706 - 10714
SHIELDS ET AL., J BIOL CHEM, vol. 277, 2002, pages 26733 - 26740
SHIELDS ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 276, no. 9, 2001, pages 6591 - 6604
SHINKAWA ET AL., J BIOL CHEM, vol. 278, 2003, pages 3466 - 3473
SMITH; CLATWORTHY, NATURE REVIEWS IMMUNOLOGY, vol. 10, 2010, pages 328 - 343
STANLEY ET AL., J. BIOL. CHEM., vol. 261, 1984, pages 13370 - 13378
STROHL, CURRENT OPINION IN BIOTECHNOLOGY, vol. 20, 2009, pages 685 - 691
TRAIL ET AL., CURR. OPIN. IMMUNOL., vol. 11, 1999, pages 584 - 588
TSURUSHITA; VASQUEZ: "Molecular Biology of B Cells", 2004, ELSEVIER SCIENCE, article "Humanization of Monoclonal Antibodies", pages: 533 - 545
UMANA ET AL., NAT BIOTECHNOL, vol. 17, 1999, pages 176 - 180
UMANA ET AL., NATURE BIOTECH., vol. 17, 1999, pages 176 - 180
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
VERI ET AL., IMMUNOLOGY, vol. 121, 2007, pages 392 - 404
WEINER ET AL., NATURE REVIEWS IMMUNOLOGY, vol. 10, 2010, pages 317 - 327
WESOLOWSKI ET AL., MED MICROBIOL IMMUNOL, vol. 198, no. 3, 2009, pages 157 - 174
WU ET AL., NATURE BIOTECHNOLOGY, vol. 25, no. 11, 2007, pages 1290 - 1297
XIE ET AL., J IMMUNOL METHODS, vol. 296, 2005, pages 95 - 101
YEUNG ET AL., J IMMUNOL, vol. 182, 2010, pages 7663 - 7671
ZEIDLER R ET AL: "The Fc-region of a new class of intact bispecific antibody mediates activation of accessory cells and NK cells and induces direct phagocytosis of tumour cells", BRITISH JOURNAL OF CANCER, vol. 83, no. 2, July 2000 (2000-07-01), pages 261 - 266, XP002610993, ISSN: 0007-0920 *
ZUO ET AL., PROTEIN ENGINEERING, vol. 13, no. 5, 2000, pages 361 - 367

Cited By (300)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8981061B2 (en) 2001-03-20 2015-03-17 Novo Nordisk A/S Receptor TREM (triggering receptor expressed on myeloid cells) and uses thereof
USRE47770E1 (en) 2002-07-18 2019-12-17 Merus N.V. Recombinant production of mixtures of antibodies
US10934571B2 (en) 2002-07-18 2021-03-02 Merus N.V. Recombinant production of mixtures of antibodies
US10603357B2 (en) 2004-11-29 2020-03-31 Bristol-Myers Squibb Company Therapeutic TREM-1 peptides
US9273111B2 (en) 2004-11-29 2016-03-01 Universite De Lorraine Therapeutic TREM-1 peptides
US9266967B2 (en) 2007-12-21 2016-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10138293B2 (en) 2007-12-21 2018-11-27 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US10927163B2 (en) 2007-12-21 2021-02-23 Hoffmann-La Roche, Inc. Bivalent, bispecific antibodies
US9382323B2 (en) 2009-04-02 2016-07-05 Roche Glycart Ag Multispecific antibodies comprising full length antibodies and single chain fab fragments
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9676845B2 (en) 2009-06-16 2017-06-13 Hoffmann-La Roche, Inc. Bispecific antigen binding proteins
US11673945B2 (en) 2009-06-16 2023-06-13 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US10640555B2 (en) 2009-06-16 2020-05-05 Hoffmann-La Roche Inc. Bispecific antigen binding proteins
US9994646B2 (en) 2009-09-16 2018-06-12 Genentech, Inc. Coiled coil and/or tether containing protein complexes and uses thereof
WO2011091078A3 (en) * 2010-01-19 2011-12-01 Xencor, Inc. Antibody fc variants with enhanced complement activity
WO2011097527A3 (en) * 2010-02-04 2012-01-26 Xencor, Inc. Immunoprotection of therapeutic moieties using enhanced fc regions
US10106600B2 (en) 2010-03-26 2018-10-23 Roche Glycart Ag Bispecific antibodies
US9879095B2 (en) 2010-08-24 2018-01-30 Hoffman-La Roche Inc. Bispecific antibodies comprising a disulfide stabilized-Fv fragment
WO2012058768A1 (en) * 2010-11-05 2012-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
US9562109B2 (en) 2010-11-05 2017-02-07 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US10875931B2 (en) 2010-11-05 2020-12-29 Zymeworks, Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US11618790B2 (en) 2010-12-23 2023-04-04 Hoffmann-La Roche Inc. Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
US10611825B2 (en) 2011-02-28 2020-04-07 Hoffmann La-Roche Inc. Monovalent antigen binding proteins
US10793621B2 (en) 2011-02-28 2020-10-06 Hoffmann-La Roche Inc. Nucleic acid encoding dual Fc antigen binding proteins
US9982036B2 (en) 2011-02-28 2018-05-29 Hoffmann-La Roche Inc. Dual FC antigen binding proteins
US10711051B2 (en) 2011-03-03 2020-07-14 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US9499605B2 (en) 2011-03-03 2016-11-22 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US10155803B2 (en) 2011-03-03 2018-12-18 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
WO2013002362A1 (en) 2011-06-30 2013-01-03 中外製薬株式会社 Heterodimerized polypeptide
EP4011913A1 (en) 2011-06-30 2022-06-15 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
WO2013004842A2 (en) 2011-07-06 2013-01-10 Genmab A/S Antibody variants and uses thereof
EP3970746A2 (en) 2011-07-06 2022-03-23 Genmab B.V. Polypeptide variants and uses thereof
US11639397B2 (en) 2011-08-23 2023-05-02 Roche Glycart Ag Bispecific antibodies specific for T-cell activating antigens and a tumor antigen and methods of use
WO2013033008A2 (en) 2011-08-26 2013-03-07 Merrimack Pharmaceuticals, Inc. Tandem fc bispecific antibodies
WO2013033008A3 (en) * 2011-08-26 2013-04-25 Merrimack Pharmaceuticals, Inc. Tandem fc bispecific antibodies
CN103857700A (en) * 2011-08-26 2014-06-11 梅里麦克制药股份有限公司 Tandem FC bispecific antibodies
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
RU2675319C2 (en) * 2011-11-04 2018-12-18 Займворкс Инк. STABLE HETERODIMERIC ANTIBODY DESIGN WITH MUTATIONS IN Fc DOMAIN
KR20140092370A (en) * 2011-11-04 2014-07-23 자임워크스 인코포레이티드 Stable heterodimeric antibody design with mutations in the fc domain
US10457742B2 (en) 2011-11-04 2019-10-29 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the Fc domain
KR102052774B1 (en) * 2011-11-04 2019-12-04 자임워크스 인코포레이티드 Stable heterodimeric antibody design with mutations in the fc domain
EP2773671A4 (en) * 2011-11-04 2015-06-03 Zymeworks Inc Stable heterodimeric antibody design with mutations in the fc domain
CN104080811B (en) * 2011-11-04 2019-09-27 酵活有限公司 There is the antibody design of the stabilization heterodimeric of mutation in Fc structural domain
US9574010B2 (en) 2011-11-04 2017-02-21 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the Fc domain
WO2013063702A1 (en) 2011-11-04 2013-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
AU2017245451B2 (en) * 2011-11-04 2019-08-22 Zymeworks Bc Inc. Stable heterodimeric antibody design with mutations in the Fc domain
AU2017245451B9 (en) * 2011-11-04 2019-08-29 Zymeworks Bc Inc. Stable heterodimeric antibody design with mutations in the Fc domain
AU2012332021B8 (en) * 2011-11-04 2017-10-12 Zymeworks Bc Inc. Stable heterodimeric antibody design with mutations in the Fc domain
AU2012332021A8 (en) * 2011-11-04 2017-10-12 Zymeworks Bc Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US9988460B2 (en) 2011-11-04 2018-06-05 Zymeworks Inc. Crystal structures of heterodimeric Fc domains
CN109897103A (en) * 2011-11-04 2019-06-18 酵活有限公司 There is the antibody design of the stabilization heterodimeric of mutation in Fc structural domain
AU2012332021B2 (en) * 2011-11-04 2017-07-20 Zymeworks Bc Inc. Stable heterodimeric antibody design with mutations in the Fc domain
RU2810540C2 (en) * 2011-11-04 2023-12-27 Займворкс Бк Инк. DEVELOPMENT OF STABLE HETERODIMERIC ANTIBODY WITH MUTATIONS IN Fc DOMAIN
CN104080811A (en) * 2011-11-04 2014-10-01 酵活有限公司 Stable heterodimeric antibody design with mutations in the Fc domain
US9732155B2 (en) 2011-11-04 2017-08-15 Zymeworks Inc. Crystal structures of heterodimeric Fc domains
US9688758B2 (en) 2012-02-10 2017-06-27 Genentech, Inc. Single-chain antibodies and other heteromultimers
US10189904B2 (en) 2012-02-15 2019-01-29 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9000127B2 (en) 2012-02-15 2015-04-07 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10906975B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation with antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US10906965B2 (en) 2012-02-15 2021-02-02 Novo Nordisk A/S Methods of treating autoimmune disease or chronic inflammation wtih antibodies that bind peptidoglycan recognition protein 1
US9663568B2 (en) 2012-02-15 2017-05-30 Novo Nordisk A/S Antibodies that bind peptidoglycan recognition protein 1
US10150809B2 (en) 2012-02-15 2018-12-11 Bristol-Myers Squibb Company Antibodies that bind peptidoglycan recognition protein 1
US9550830B2 (en) 2012-02-15 2017-01-24 Novo Nordisk A/S Antibodies that bind and block triggering receptor expressed on myeloid cells-1 (TREM-1)
US9714292B2 (en) 2012-04-05 2017-07-25 Hoffmann-La Roche Inc. Bispecific antibodies against human TWEAK and human IL17 and uses thereof
WO2013150043A1 (en) 2012-04-05 2013-10-10 F. Hoffmann-La Roche Ag Bispecific antibodies against human tweak and human il17 and uses thereof
US11926859B2 (en) 2012-04-20 2024-03-12 Merus N.V. Methods and means for the production of Ig-like molecules
US9358286B2 (en) 2012-04-20 2016-06-07 Merus B.V. Methods and means for the production of IG-like molecules
US10329596B2 (en) 2012-04-20 2019-06-25 Merus N.V. Methods and means for the production of Ig-like molecules
US10752929B2 (en) 2012-04-20 2020-08-25 Merus N.V. Methods and means for the production of ig-like molecules
US10337045B2 (en) 2012-04-20 2019-07-02 Merus N.V. Methods and means for the production of Ig-like molecules
US9248182B2 (en) 2012-04-20 2016-02-02 Merus B.V. Methods and means for the production of Ig-like molecules
US9758805B2 (en) 2012-04-20 2017-09-12 Merus N.V. Methods and means for the production of Ig-like molecules
US9248181B2 (en) 2012-04-20 2016-02-02 Merus B.V. Methods and means for the production of Ig-like molecules
EP4310191A2 (en) 2012-06-14 2024-01-24 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified fc region
WO2013187495A1 (en) 2012-06-14 2013-12-19 中外製薬株式会社 ANTIGEN-BINDING MOLECULE CONTAINING MODIFIED Fc REGION
US10508154B2 (en) 2012-06-25 2019-12-17 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
US11421022B2 (en) 2012-06-27 2022-08-23 Hoffmann-La Roche Inc. Method for making antibody Fc-region conjugates comprising at least one binding entity that specifically binds to a target and uses thereof
US10106612B2 (en) 2012-06-27 2018-10-23 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
US11407836B2 (en) 2012-06-27 2022-08-09 Hoffmann-La Roche Inc. Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
EP3632462A1 (en) 2012-07-06 2020-04-08 Genmab B.V. Dimeric protein with triple mutations
WO2014006217A1 (en) 2012-07-06 2014-01-09 Genmab B.V. Dimeric protein with triple mutations
US11248037B2 (en) 2012-07-13 2022-02-15 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
US10358479B2 (en) 2012-07-13 2019-07-23 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
EP3721900A1 (en) 2012-08-24 2020-10-14 Chugai Seiyaku Kabushiki Kaisha Fcgammariib-specific fc region variant
WO2014030728A1 (en) 2012-08-24 2014-02-27 中外製薬株式会社 Fcγriib-specific fc region variant
KR102390177B1 (en) * 2012-09-27 2022-04-26 메뤼스 엔.페. Bispecific igg antibodies as t cell engagers
EP3470431A1 (en) * 2012-09-27 2019-04-17 Merus N.V. Bispecific igg antibodies as t cell engagers
US10358492B2 (en) 2012-09-27 2019-07-23 Merus N.V. Bispecific IgG antibodies as T cell engagers
WO2014051433A1 (en) * 2012-09-27 2014-04-03 Merus B.V. BISPECIFIC IgG ANTIBODIES AS T CELL ENGAGERS
KR20150076178A (en) * 2012-09-27 2015-07-06 메뤼스 베.페. Bispecific igg antibodies as t cell engagers
US9771573B2 (en) 2012-10-03 2017-09-26 Zymeworks Inc. Methods of quantitating heavy and light chain polypeptide pairs
US10087250B2 (en) 2012-10-08 2018-10-02 Roche Glycart Ag Fc-free antibodies comprising two fab-fragments and methods of use
US11286293B2 (en) 2012-11-28 2022-03-29 Zymeworks, Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US11078296B2 (en) 2012-11-28 2021-08-03 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US10077298B2 (en) 2012-11-28 2018-09-18 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
WO2014104165A1 (en) 2012-12-27 2014-07-03 中外製薬株式会社 Heterodimerized polypeptide
WO2014108198A1 (en) 2013-01-10 2014-07-17 Genmab B.V. Human igg1 fc region variants and uses thereof
WO2014108483A1 (en) 2013-01-10 2014-07-17 Genmab B.V. Inert format
EP3738979A1 (en) 2013-01-10 2020-11-18 Genmab A/S Inert format
US11718667B2 (en) 2013-01-14 2023-08-08 Xencor, Inc. Optimized antibody variable regions
US10738133B2 (en) 2013-01-14 2020-08-11 Xencor, Inc. Heterodimeric proteins
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10472427B2 (en) 2013-01-14 2019-11-12 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US11634506B2 (en) 2013-01-14 2023-04-25 Xencor, Inc. Heterodimeric proteins
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
US10851171B2 (en) 2013-02-05 2020-12-01 Engmab Sarl Method for the selection of antibodies against BCMA
US10077315B2 (en) 2013-02-05 2018-09-18 Engmab Sàrl Bispecific antibodies against CD3 and BCMA
US9963513B2 (en) 2013-02-05 2018-05-08 Engmab Sàrl Method for the selection of antibodies against BCMA
US10781257B2 (en) 2013-02-26 2020-09-22 Roche GlyeArt AG Bispecific T cell activating antigen binding molecules
US10155815B2 (en) 2013-02-26 2018-12-18 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US11459404B2 (en) 2013-02-26 2022-10-04 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
US10781258B2 (en) 2013-02-26 2020-09-22 Roche Glycart Ag Bispecific T cell activating antigen binding molecules
CN105705519A (en) * 2013-03-06 2016-06-22 梅里麦克制药股份有限公司 Anti-C-MET tandem Fc bispecific antibodies
US9458245B2 (en) 2013-03-06 2016-10-04 Merrimack Pharmaceuticals, Inc. ANTI-C-MET tandem Fc bispecific antibodies
WO2014138449A1 (en) 2013-03-06 2014-09-12 Merrimack Pharmaceuticals, Inc. Anti-c-met tandem fc bispecific antibodies
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
EP3486255A3 (en) * 2013-03-13 2019-11-13 Ibentrus Inc. Protein in which electrical interaction is introduced within hydrophobic interaction site and preparation method therefor
US11299554B2 (en) 2013-03-15 2022-04-12 Xencor, Inc. Heterodimeric proteins
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
US11814423B2 (en) 2013-03-15 2023-11-14 Xencor, Inc. Heterodimeric proteins
US10287364B2 (en) 2013-03-15 2019-05-14 Xencor, Inc. Heterodimeric proteins
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10544187B2 (en) 2013-03-15 2020-01-28 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
WO2015001085A1 (en) 2013-07-05 2015-01-08 Genmab B.V. Humanized or chimeric cd3 antibodies
EP3693385A1 (en) 2013-07-05 2020-08-12 Genmab A/S Humanized or chimeric cd3 antibodies
WO2015048272A1 (en) * 2013-09-25 2015-04-02 Amgen Inc. V-c-fc-v-c antibody
US10323099B2 (en) 2013-10-11 2019-06-18 Hoffmann-La Roche Inc. Multispecific domain exchanged common variable light chain antibodies
WO2015068847A1 (en) 2013-11-11 2015-05-14 中外製薬株式会社 Antigen-binding molecule containing modified antibody variable region
US11325981B2 (en) 2013-11-27 2022-05-10 Zymeworks Inc. Bispecific antigen-binding constructs targeting Her2
US10947319B2 (en) 2013-11-27 2021-03-16 Zymeworks Inc. Bispecific antigen-binding constructs targeting HER2
US11186650B2 (en) 2013-12-17 2021-11-30 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10640572B2 (en) 2013-12-17 2020-05-05 Genentech, Inc. Anti-CD3 antibodies and methods of use
US11732054B2 (en) 2013-12-17 2023-08-22 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10174124B2 (en) 2013-12-17 2019-01-08 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10865251B2 (en) 2013-12-17 2020-12-15 Genentech, Inc. Anti-CD3 antibodies and methods of use
US11530275B2 (en) 2013-12-17 2022-12-20 Genentech, Inc. Anti-CD3 antibodies and methods of use
WO2015104346A1 (en) 2014-01-09 2015-07-16 Genmab B.V. Humanized or chimeric cd3 antibodies
US10844127B2 (en) 2014-02-28 2020-11-24 Merus N.V. Antibodies that bind EGFR and ErbB3
US11820825B2 (en) 2014-02-28 2023-11-21 Merus N.V. Methods of treating a subject having an EGFR-positive and/or ErbB-3-positive tumor
US11279770B2 (en) 2014-02-28 2022-03-22 Merus N.V. Antibody that binds ErbB-2 and ErbB-3
US11840579B2 (en) 2014-03-28 2023-12-12 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US10858451B2 (en) 2014-03-28 2020-12-08 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US9822186B2 (en) 2014-03-28 2017-11-21 Xencor, Inc. Bispecific antibodies that bind to CD38 and CD3
US11306156B2 (en) 2014-05-28 2022-04-19 Zymeworks Inc. Modified antigen binding polypeptide constructs and uses thereof
WO2016005593A1 (en) 2014-07-11 2016-01-14 Genmab A/S Antibodies binding axl
EP3763738A1 (en) 2014-07-11 2021-01-13 Genmab A/S Antibodies binding axl
US10179814B2 (en) 2014-07-17 2019-01-15 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
US11072654B2 (en) 2014-07-17 2021-07-27 Novo Nordisk A/S Site directed mutagenesis of TREM-1 antibodies for decreasing viscosity
US10611840B2 (en) 2014-08-04 2020-04-07 Hoffman-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11117965B2 (en) 2014-08-04 2021-09-14 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10611841B2 (en) 2014-08-04 2020-04-07 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US9914776B2 (en) 2014-08-04 2018-03-13 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US11084877B2 (en) 2014-09-12 2021-08-10 Genentech, Inc. Anti-CLL-1 antibodies and immunoconjugates
CN107108720A (en) * 2014-11-06 2017-08-29 豪夫迈·罗氏有限公司 Fc region variants and its application method that FCRN with change is combined
WO2016071376A3 (en) * 2014-11-06 2016-06-30 F. Hoffmann-La Roche Ag Fc-region variants with modified fcrn-binding and methods of use
US11440971B2 (en) 2014-11-06 2022-09-13 Hoffmann-La Roche Inc. Fc-region variants with modified FcRn-binding and methods of use
EP3611188A1 (en) * 2014-11-06 2020-02-19 F. Hoffmann-La Roche AG Fc-region variants with modified fcrn-binding and methods of use
US10683368B2 (en) 2014-11-06 2020-06-16 Hoffmann-La Roche Inc. Fc-region variants with modified FcRn-binding and methods of use
US11613587B2 (en) 2014-11-20 2023-03-28 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
US10781262B2 (en) 2014-11-20 2020-09-22 Hoffmann-La Roche Inc. Combination therapy of T cell activating bispecific antigen binding molecules and PD-1 axis binding antagonists
US10913803B2 (en) 2014-11-26 2021-02-09 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11673972B2 (en) 2014-11-26 2023-06-13 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11352442B2 (en) 2014-11-26 2022-06-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US11225528B2 (en) 2014-11-26 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11111315B2 (en) 2014-11-26 2021-09-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US11859011B2 (en) 2014-11-26 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10526417B2 (en) 2014-11-26 2020-01-07 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US9856327B2 (en) 2014-11-26 2018-01-02 Xencor, Inc. Heterodimeric antibodies to CD3 X CD123
US11945880B2 (en) 2014-11-26 2024-04-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10889653B2 (en) 2014-11-26 2021-01-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US9850320B2 (en) 2014-11-26 2017-12-26 Xencor, Inc. Heterodimeric antibodies to CD3 X CD20
US10633457B2 (en) 2014-12-03 2020-04-28 Hoffmann-La Roche Inc. Multispecific antibodies
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
US10544220B2 (en) 2015-01-08 2020-01-28 Genmab A/S Bispecific antibodies against CD3 and CD20
US10774156B2 (en) 2015-01-22 2020-09-15 Eli Lilly And Company IgG bispecific antibodies and processes for preparation
US11091548B2 (en) 2015-03-05 2021-08-17 Xencor, Inc. Modulation of T cells with bispecific antibodies and Fc fusions
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
US11192950B2 (en) 2015-06-16 2021-12-07 Genentech, Inc. Humanized and affinity matured antibodies to FcRH5 and methods of use
US10501545B2 (en) 2015-06-16 2019-12-10 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
US11466087B2 (en) 2015-06-16 2022-10-11 Genentech, Inc. Anti-CLL-1 antibodies and methods of use
US10323094B2 (en) 2015-06-16 2019-06-18 Genentech, Inc. Humanized and affinity matured antibodies to FcRH5 and methods of use
US11007267B2 (en) 2015-06-16 2021-05-18 Genentech, Inc. Anti-CD3 antibodies and methods of use
US10266593B2 (en) 2015-07-10 2019-04-23 Merus N.V. Human CD3 binding antibody
US11739148B2 (en) 2015-07-10 2023-08-29 Merus N.V. Human CD3 binding antibody
EP3730520A1 (en) 2015-07-10 2020-10-28 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
US9914777B2 (en) 2015-07-10 2018-03-13 Merus N.V. Human CD3 binding antibody
WO2017009258A1 (en) 2015-07-10 2017-01-19 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
US11359015B2 (en) 2015-07-15 2022-06-14 Genmab A/S Humanized or chimeric CD3 antibodies
US11286300B2 (en) 2015-10-01 2022-03-29 Hoffmann-La Roche Inc. Humanized anti-human CD19 antibodies and methods of use
US10766967B2 (en) 2015-10-02 2020-09-08 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
WO2017066878A1 (en) * 2015-10-22 2017-04-27 Canadian Blood Services Monovalent chimeras
US11939394B2 (en) 2015-10-23 2024-03-26 Merus N.V. Binding molecules that inhibit cancer growth
WO2017086367A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Combination therapy using t cell redirection antigen binding molecule against cell having immunosuppressing function
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
US11623957B2 (en) 2015-12-07 2023-04-11 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
US11013801B2 (en) 2015-12-09 2021-05-25 Hoffmann-La Roche Inc. Treatment method
US10596257B2 (en) 2016-01-08 2020-03-24 Hoffmann-La Roche Inc. Methods of treating CEA-positive cancers using PD-1 axis binding antagonists and anti-CEA/anti-CD3 bispecific antibodies
WO2017121877A1 (en) 2016-01-13 2017-07-20 Genmab A/S Axl-specific antibody-drug conjugates for cancer treatment
EP3851457A1 (en) 2016-01-21 2021-07-21 Novartis AG Multispecific molecules targeting cll-1
WO2017125897A1 (en) 2016-01-21 2017-07-27 Novartis Ag Multispecific molecules targeting cll-1
WO2017159287A1 (en) 2016-03-14 2017-09-21 中外製薬株式会社 Cell injury inducing therapeutic drug for use in cancer therapy
US11242390B2 (en) 2016-03-22 2022-02-08 Hoffmann-La Roche Inc. Protease-activated T cell bispecific molecules
US11236170B2 (en) 2016-06-14 2022-02-01 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11492407B2 (en) 2016-06-14 2022-11-08 Xencor, Inc. Bispecific checkpoint inhibitor antibodies
US11225521B2 (en) 2016-06-28 2022-01-18 Xencor, Inc. Heterodimeric antibodies that bind somatostatin receptor 2
WO2018011421A1 (en) 2016-07-14 2018-01-18 Genmab A/S Multispecific antibodies against cd40 and cd137
US10882918B2 (en) 2016-09-30 2021-01-05 Hoffmann-La Roche Inc. Bispecific T cell activating antigen binding molecules
US10550185B2 (en) 2016-10-14 2020-02-04 Xencor, Inc. Bispecific heterodimeric fusion proteins containing IL-15-IL-15Rα Fc-fusion proteins and PD-1 antibody fragments
US10501543B2 (en) 2016-10-14 2019-12-10 Xencor, Inc. IL15/IL15Rα heterodimeric Fc-fusion proteins
WO2018083126A1 (en) 2016-11-01 2018-05-11 Genmab B.V. Polypeptide variants and uses thereof
US11466094B2 (en) 2016-11-15 2022-10-11 Genentech, Inc. Dosing for treatment with anti-CD20/anti-CD3 bispecific antibodies
WO2018146317A1 (en) 2017-02-10 2018-08-16 Genmab B.V. Polypeptide variants and uses thereof
WO2018162749A1 (en) 2017-03-09 2018-09-13 Genmab A/S Antibodies against pd-l1
US11780925B2 (en) 2017-03-31 2023-10-10 Merus N.V. ErbB-2 and ErbB3 binding bispecific antibodies for use in the treatment of cells that have an NRG1 fusion gene
WO2018178396A1 (en) 2017-03-31 2018-10-04 Genmab Holding B.V. Bispecific anti-cd37 antibodies, monoclonal anti-cd37 antibodies and methods of use thereof
WO2018201051A1 (en) 2017-04-28 2018-11-01 Novartis Ag Bcma-targeting agent, and combination therapy with a gamma secretase inhibitor
WO2018224609A1 (en) 2017-06-07 2018-12-13 Genmab B.V. Therapeutic antibodies based on mutated igg hexamers
US11084863B2 (en) 2017-06-30 2021-08-10 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15 IL-15alpha and antigen binding domains
WO2019025545A1 (en) 2017-08-04 2019-02-07 Genmab A/S Binding agents binding to pd-l1 and cd137 and use thereof
WO2019032624A1 (en) * 2017-08-08 2019-02-14 Pionyr Immunotherapeutics, Inc. Compositions and methods for disabling meyloid cells expressing trem1
US11773170B2 (en) 2017-08-09 2023-10-03 Merus N.V. Antibodies that bind EGFR and cMET
US11312770B2 (en) 2017-11-08 2022-04-26 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-PD-1 sequences
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US11319355B2 (en) 2017-12-19 2022-05-03 Xencor, Inc. Engineered IL-2 Fc fusion proteins
WO2019131988A1 (en) 2017-12-28 2019-07-04 Chugai Seiyaku Kabushiki Kaisha Cytotoxicity-inducing therapeutic agent
WO2019145455A1 (en) 2018-01-24 2019-08-01 Genmab B.V. Polypeptide variants and uses thereof
US11866498B2 (en) 2018-02-08 2024-01-09 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
WO2019155008A1 (en) 2018-02-09 2019-08-15 Genmab A/S Pharmaceutical compositions comprising bispecific antibodies directed against cd3 and cd20 and their uses
WO2019175198A2 (en) 2018-03-12 2019-09-19 Genmab A/S Antibodies
US11155618B2 (en) 2018-04-02 2021-10-26 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
US11919954B2 (en) 2018-04-02 2024-03-05 Bristol-Myers Squibb Company Anti-TREM-1 antibodies and uses thereof
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
US11524991B2 (en) 2018-04-18 2022-12-13 Xencor, Inc. PD-1 targeted heterodimeric fusion proteins containing IL-15/IL-15Ra Fc-fusion proteins and PD-1 antigen binding domains and uses thereof
US11505595B2 (en) 2018-04-18 2022-11-22 Xencor, Inc. TIM-3 targeted heterodimeric fusion proteins containing IL-15/IL-15RA Fc-fusion proteins and TIM-3 antigen binding domains
WO2019211472A1 (en) 2018-05-03 2019-11-07 Genmab B.V. Antibody variant combinations and uses thereof
WO2019232484A1 (en) 2018-06-01 2019-12-05 Compugen Ltd Anti-pvrig/anti-tigit bispecific antibodies and methods of use
WO2019243636A1 (en) 2018-06-22 2019-12-26 Genmab Holding B.V. Anti-cd37 antibodies and anti-cd20 antibodies, compositions and methods of use thereof
WO2020012038A1 (en) 2018-07-13 2020-01-16 Genmab A/S Trogocytosis-mediated therapy using cd38 antibodies
WO2020012036A1 (en) 2018-07-13 2020-01-16 Genmab A/S Variants of cd38 antibody and uses thereof
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
WO2020070313A1 (en) 2018-10-04 2020-04-09 Genmab Holding B.V. Pharmaceutical compositions comprising bispecific anti-cd37 antibodies
WO2020094744A1 (en) 2018-11-06 2020-05-14 Genmab A/S Antibody formulation
US10836828B2 (en) 2019-02-06 2020-11-17 Pionyr Immunotherapeutics, Inc. Anti-TREM1 antibodies and related methods
US11472890B2 (en) 2019-03-01 2022-10-18 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
WO2020225456A1 (en) 2019-05-09 2020-11-12 Genmab B.V. Dosage regimens for a combination of anti-dr5 antibodies for use in treating cancer
WO2020246563A1 (en) 2019-06-05 2020-12-10 中外製薬株式会社 Antibody cleavage site-binding molecule
WO2021006328A1 (en) 2019-07-10 2021-01-14 Chugai Seiyaku Kabushiki Kaisha Claudin-6 binding molecules and uses thereof
WO2021028587A1 (en) 2019-08-15 2021-02-18 Genmab A/S Pharmaceutical compositions comprising bispecific antibodies directed against cd3 and cd20 and their uses
WO2021089850A1 (en) 2019-11-06 2021-05-14 Genmab B.V. Antibody variant combinations and uses thereof
US11845799B2 (en) 2019-12-13 2023-12-19 Genentech, Inc. Anti-Ly6G6D antibodies and methods of use
WO2021131021A1 (en) 2019-12-27 2021-07-01 中外製薬株式会社 Anti-ctla-4 antibody and use thereof
WO2021144457A1 (en) 2020-01-16 2021-07-22 Genmab A/S Formulations of cd38 antibodies and uses thereof
WO2021156258A1 (en) 2020-02-04 2021-08-12 BioNTech SE Treatment involving antigen vaccination and binding agents binding to pd-l1 and cd137
WO2021155916A1 (en) 2020-02-04 2021-08-12 BioNTech SE Treatment involving antigen vaccination and binding agents binding to pd-l1 and cd137
KR20220142441A (en) 2020-02-14 2022-10-21 쿄와 기린 가부시키가이샤 Bispecific antibody that binds to CD3
US11261254B1 (en) 2020-03-18 2022-03-01 Genmab A/S Antibodies
WO2021185934A1 (en) 2020-03-18 2021-09-23 Genmab A/S Antibodies binding to b7h4
US11952423B2 (en) 2020-03-30 2024-04-09 Mie University Bispecific antibody
WO2021200857A1 (en) 2020-03-30 2021-10-07 国立大学法人三重大学 Bispecific antibody
KR20220160598A (en) 2020-03-30 2022-12-06 고쿠리츠다이가쿠호진 미에다이가쿠 bispecific antibody
WO2021201087A1 (en) 2020-03-31 2021-10-07 Chugai Seiyaku Kabushiki Kaisha Method for producing multispecific antigen-binding molecules
WO2021224499A1 (en) 2020-05-08 2021-11-11 Genmab A/S Bispecific antibodies against cd3 and cd20
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
US11780920B2 (en) 2020-06-19 2023-10-10 Hoffmann-La Roche Inc. Antibodies binding to CD3 and CD19
WO2022013787A1 (en) 2020-07-16 2022-01-20 Novartis Ag Anti-betacellulin antibodies, fragments thereof, and multi-specific binding molecules
WO2022018294A1 (en) 2020-07-23 2022-01-27 Genmab B.V. A combination of anti-dr5 antibodies and an immunomodulatory imide drug for use in treating multiple myeloma
WO2022025220A1 (en) 2020-07-31 2022-02-03 中外製薬株式会社 Pharmaceutical composition including cell expressing chimeric receptor
WO2022029011A1 (en) 2020-08-06 2022-02-10 BioNTech SE Binding agents for coronavirus s protein
US11591401B2 (en) 2020-08-19 2023-02-28 Xencor, Inc. Anti-CD28 compositions
US11919958B2 (en) 2020-08-19 2024-03-05 Xencor, Inc. Anti-CD28 compositions
WO2022045276A1 (en) 2020-08-28 2022-03-03 中外製薬株式会社 Heterodimer fc polypeptide
WO2022049220A2 (en) 2020-09-02 2022-03-10 Genmab A/S Antibody therapy
US11608383B2 (en) 2020-09-10 2023-03-21 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating follicular lymphoma
US11548952B2 (en) 2020-09-10 2023-01-10 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating diffuse large B-cell lymphoma
US11535679B2 (en) 2020-09-10 2022-12-27 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating follicular lymphoma
US11845805B2 (en) 2020-09-10 2023-12-19 Genmab A/S Bispecific antibody against CD3 and CD20 in combination therapy for treating diffuse large B-cell lymphoma
US11858995B2 (en) 2020-09-10 2024-01-02 Genmab A/S Bispecific antibodies against CD3 and CD20 for treating chronic lymphocytic leukemia
WO2022069724A1 (en) 2020-10-02 2022-04-07 Genmab A/S Antibodies capable of binding to ror2 and bispecific antibodies binding to ror2 and cd3
WO2022097065A2 (en) 2020-11-06 2022-05-12 Novartis Ag ANTIBODY Fc VARIANTS
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
US11859012B2 (en) 2021-03-10 2024-01-02 Xencor, Inc. Heterodimeric antibodies that bind CD3 and GPC3
WO2022189667A1 (en) 2021-03-12 2022-09-15 Genmab A/S Non-activating antibody variants
WO2022234146A1 (en) 2021-05-07 2022-11-10 Genmab A/S PHARMACEUTICAL COMPOSITIONS COMPRISING BISPECIFIC ANTIBODIES BINDING TO B7H4 and CD3
WO2022268740A1 (en) 2021-06-21 2022-12-29 Genmab A/S Combination dosage regime of cd137 and pd-l1 binding agents
WO2022270612A1 (en) 2021-06-25 2022-12-29 中外製薬株式会社 Use of anti-ctla-4 antibody
WO2022270611A1 (en) 2021-06-25 2022-12-29 中外製薬株式会社 Anti–ctla-4 antibody
WO2023031473A1 (en) 2021-09-06 2023-03-09 Genmab B.V. Antibodies capable of binding to cd27, variants thereof and uses thereof
US11952420B2 (en) 2021-09-10 2024-04-09 Bristol-Myers Squibb Company Nucleic acids encoding anti-TREM-1 antibodies
WO2023057571A1 (en) 2021-10-08 2023-04-13 Genmab A/S Antibodies binding to cd30 and cd3
WO2023174952A1 (en) 2022-03-15 2023-09-21 Genmab A/S Binding agents binding to epcam and/or cd137
WO2023174521A1 (en) 2022-03-15 2023-09-21 Genmab A/S Binding agents binding to epcam and cd137
WO2023198839A2 (en) 2022-04-13 2023-10-19 Genmab A/S Bispecific antibodies against cd3 and cd20
WO2023209568A1 (en) 2022-04-26 2023-11-02 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2023218046A1 (en) 2022-05-12 2023-11-16 Genmab A/S Binding agents capable of binding to cd27 in combination therapy
WO2023218051A1 (en) 2022-05-12 2023-11-16 Genmab A/S Binding agents capable of binding to cd27 in combination therapy

Also Published As

Publication number Publication date
US20200123274A1 (en) 2020-04-23
US20110054151A1 (en) 2011-03-03
US9493578B2 (en) 2016-11-15
US11401348B2 (en) 2022-08-02
US20170058053A1 (en) 2017-03-02

Similar Documents

Publication Publication Date Title
US11401348B2 (en) Heterodimeric Fc variants
US20200223939A1 (en) Multispecific antibody analogs comprising a common light chain, and methods of their preparation and use
US11814423B2 (en) Heterodimeric proteins
US11634506B2 (en) Heterodimeric proteins
US20140377269A1 (en) Multivalent antibody analogs, and methods of their preparation and use
CA2902739C (en) Heterodimeric proteins
US20180215834A1 (en) Novel heterodimeric proteins
US8362210B2 (en) Antibody variants with enhanced complement activity
AU2018200008A1 (en) Modulation of T Cells with Bispecific Antibodies and FC Fusions

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10752696

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10752696

Country of ref document: EP

Kind code of ref document: A1